13,270 results on '"Vesicular Transport Proteins"'
Search Results
102. Researchers from Mukogawa Women's University Report on Findings in Influenza (Soybean Extract Inhibits Influenza Virus Entry: Mechanistic Insights).
- Abstract
Researchers from Mukogawa Women's University in Hyogo, Japan have discovered that soybean extract has inhibitory effects on the entry of influenza viruses into cells. The study found that soybean extract hindered virus uptake by cells and targeted the clathrin-dependent endocytosis pathway. These findings suggest that soybean extract could be a potential innovative antiviral intervention for influenza. The research was supported by Grants-in-Aid for Scientific Research. [Extracted from the article]
- Published
- 2024
103. Department of Food Science and Nutrition Researcher Targets Muscular Dystrophy (Caveolin and NOS in the Development of Muscular Dystrophy).
- Abstract
A recent study conducted by researchers at the Department of Food Science and Nutrition at Nara Women's University in Japan explores the role of caveolin and nitric oxide synthase (NOS) in the development of muscular dystrophy. Caveolin is a structural protein involved in molecular transport and intercellular interactions within cells. Mutations in caveolin-3 have been linked to limb-girdle muscular dystrophy, and altered expression of caveolin-3 has been detected in Duchenne muscular dystrophy. The study suggests that NOS may also play a role in the pathology of muscular dystrophy and discusses potential treatment strategies. [Extracted from the article]
- Published
- 2024
104. The driver landscape of sporadic chordoma.
- Author
-
Tarpey, Patrick S, Behjati, Sam, Young, Matthew D, Martincorena, Inigo, Alexandrov, Ludmil B, Farndon, Sarah J, Guzzo, Charlotte, Hardy, Claire, Latimer, Calli, Butler, Adam P, Teague, Jon W, Shlien, Adam, Futreal, P Andrew, Shah, Sohrab, Bashashati, Ali, Jamshidi, Farzad, Nielsen, Torsten O, Huntsman, David, Baumhoer, Daniel, Brandner, Sebastian, Wunder, Jay, Dickson, Brendan, Cogswell, Patricia, Sommer, Josh, Phillips, Joanna J, Amary, M Fernanda, Tirabosco, Roberto, Pillay, Nischalan, Yip, Stephen, Stratton, Michael R, Flanagan, Adrienne M, and Campbell, Peter J
- Subjects
Cell Line ,Tumor ,Humans ,Chordoma ,Bone Neoplasms ,T-Box Domain Proteins ,Fetal Proteins ,Vesicular Transport Proteins ,Case-Control Studies ,Gene Duplication ,Mutation ,Polymorphism ,Single Nucleotide ,Phosphatidylinositol 3-Kinases ,Class I Phosphatidylinositol 3-Kinases ,Cell Line ,Tumor ,Polymorphism ,Single Nucleotide - Abstract
Chordoma is a malignant, often incurable bone tumour showing notochordal differentiation. Here, we defined the somatic driver landscape of 104 cases of sporadic chordoma. We reveal somatic duplications of the notochordal transcription factor brachyury (T) in up to 27% of cases. These variants recapitulate the rearrangement architecture of the pathogenic germline duplications of T that underlie familial chordoma. In addition, we find potentially clinically actionable PI3K signalling mutations in 16% of cases. Intriguingly, one of the most frequently altered genes, mutated exclusively by inactivating mutation, was LYST (10%), which may represent a novel cancer gene in chordoma.Chordoma is a rare often incurable malignant bone tumour. Here, the authors investigate driver mutations of sporadic chordoma in 104 cases, revealing duplications in notochordal transcription factor brachyury (T), PI3K signalling mutations, and mutations in LYST, a potential novel cancer gene in chordoma.
- Published
- 2017
105. Rewiring a Rab regulatory network reveals a possible inhibitory role for the vesicle tether, Uso1
- Author
-
Yuan, Hua, Davis, Saralin, Ferro-Novick, Susan, and Novick, Peter
- Subjects
Rare Diseases ,Cell Membrane ,Endoplasmic Reticulum ,Golgi Apparatus ,Guanine Nucleotide Exchange Factors ,Protein Binding ,Saccharomyces cerevisiae ,Saccharomyces cerevisiae Proteins ,Vesicular Transport Proteins ,rab GTP-Binding Proteins ,membrane traffic ,Rab GTPase ,vesicle tether - Abstract
Ypt1 and Sec4 are essential Rab GTPases that control the early and late stages of the yeast secretory pathway, respectively. A chimera consisting of Ypt1 with the switch I domain of Sec4, Ypt1-SW1Sec4, is efficiently activated in vitro by the Sec4 exchange factor, Sec2. This should lead to its ectopic activation in vivo and thereby disrupt membrane traffic. Nonetheless early studies found that yeast expressing Ypt1-SW1Sec4 as the sole copy of YPT1 exhibit no growth defect. To resolve this conundrum, we have analyzed yeast expressing various levels of Ypt1-SW1Sec4 We show that even normal expression of Ypt1-SW1Sec4 leads to kinetic transport defects at a late stage of the pathway, with secretory vesicles accumulating near exocytic sites. Higher levels are toxic. Toxicity is suppressed by truncation of Uso1, a vesicle tether required for endoplasmic reticulum-Golgi traffic. The globular head of Uso1 binds to Ypt1 and its coiled-coil tail binds to the Golgi-associated SNARE, Sed5. We propose that when Uso1 is inappropriately recruited to secretory vesicles by Ypt1-SW1Sec4, the extended coiled-coil tail blocks docking to the plasma membrane. This putative inhibitory function could serve to increase the fidelity of vesicle docking.
- Published
- 2017
106. TFG facilitates outer coat disassembly on COPII transport carriers to promote tethering and fusion with ER–Golgi intermediate compartments
- Author
-
Hanna, Michael G, Block, Samuel, Frankel, EB, Hou, Feng, Johnson, Adam, Yuan, Lin, Knight, Gavin, Moresco, James J, Yates, John R, Ashton, Randolph, Schekman, Randy, Tong, Yufeng, and Audhya, Anjon
- Subjects
Underpinning research ,1.1 Normal biological development and functioning ,Generic health relevance ,Animals ,Biological Transport ,COP-Coated Vesicles ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins ,Carrier Proteins ,Endoplasmic Reticulum ,Female ,Golgi Apparatus ,Male ,Protein Transport ,Rats ,Sprague-Dawley ,Vesicular Transport Proteins ,COPII ,Trk-fused gene ,tether ,coat disassembly ,endoplasmic reticulum - Abstract
The conserved coat protein complex II (COPII) mediates the initial steps of secretory protein trafficking by assembling onto subdomains of the endoplasmic reticulum (ER) in two layers to generate cargo-laden transport carriers that ultimately fuse with an adjacent ER-Golgi intermediate compartment (ERGIC). Here, we demonstrate that Trk-fused gene (TFG) binds directly to the inner layer of the COPII coat. Specifically, the TFG C terminus interacts with Sec23 through a shared interface with the outer COPII coat and the cargo receptor Tango1/cTAGE5. Our findings indicate that TFG binding to Sec23 outcompetes these other associations in a concentration-dependent manner and ultimately promotes outer coat dissociation. Additionally, we demonstrate that TFG tethers vesicles harboring the inner COPII coat, which contributes to their clustering between the ER and ERGIC in cells. Together, our studies define a mechanism by which COPII transport carriers are retained locally at the ER/ERGIC interface after outer coat disassembly, which is a prerequisite for fusion with ERGIC membranes.
- Published
- 2017
107. Remodeling of ER‐exit sites initiates a membrane supply pathway for autophagosome biogenesis
- Author
-
Ge, Liang, Zhang, Min, Kenny, Samuel J, Liu, Dawei, Maeda, Miharu, Saito, Kota, Mathur, Anandita, Xu, Ke, and Schekman, Randy
- Subjects
Generic health relevance ,Antigens ,Neoplasm ,Autophagosomes ,Autophagy ,Autophagy-Related Proteins ,COP-Coated Vesicles ,Endoplasmic Reticulum ,Golgi Apparatus ,HeLa Cells ,Humans ,Membrane Proteins ,Membranes ,Microscopy ,Microtubule-Associated Proteins ,Neoplasm Proteins ,Organelle Biogenesis ,Protein Transport ,Protein-Tyrosine Kinases ,Vesicular Transport Proteins ,autophagosome ,autophagy ,COPII ,ER-exit sites ,FIP200 ,Hela Cells ,COPII ,ER‐exit sites ,Biochemistry and Cell Biology ,Developmental Biology - Abstract
Autophagosomes are double-membrane vesicles generated during autophagy. Biogenesis of the autophagosome requires membrane acquisition from intracellular compartments, the mechanisms of which are unclear. We previously found that a relocation of COPII machinery to the ER-Golgi intermediate compartment (ERGIC) generates ERGIC-derived COPII vesicles which serve as a membrane precursor for the lipidation of LC3, a key membrane component of the autophagosome. Here we employed super-resolution microscopy to show that starvation induces the enlargement of ER-exit sites (ERES) positive for the COPII activator, SEC12, and the remodeled ERES patches along the ERGIC A SEC12 binding protein, CTAGE5, is required for the enlargement of ERES, SEC12 relocation to the ERGIC, and modulates autophagosome biogenesis. Moreover, FIP200, a subunit of the ULK protein kinase complex, facilitates the starvation-induced enlargement of ERES independent of the other subunits of this complex and associates via its C-terminal domain with SEC12. Our data indicate a pathway wherein FIP200 and CTAGE5 facilitate starvation-induced remodeling of the ERES, a prerequisite for the production of COPII vesicles budded from the ERGIC that contribute to autophagosome formation.
- Published
- 2017
108. Activation of Parathyroid Hormone 2 Receptor Induces Decorin Expression and Promotes Wound Repair
- Author
-
Sato, Emi, Zhang, Ling-juan, Dorschner, Robert A, Adase, Christopher A, Choudhury, Biswa P, and Gallo, Richard L
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Development of treatments and therapeutic interventions ,1.1 Normal biological development and functioning ,Underpinning research ,5.2 Cellular and gene therapies ,Skin ,Animals ,Cell Differentiation ,Cells ,Cultured ,Decorin ,Disease Models ,Animal ,Female ,Gene Expression Regulation ,Immunoblotting ,Mice ,Mice ,Inbred C57BL ,Nuclear Proteins ,RNA ,RNA Splicing Factors ,RNA-Binding Proteins ,Real-Time Polymerase Chain Reaction ,Receptor ,Parathyroid Hormone ,Type 2 ,Signal Transduction ,Vesicular Transport Proteins ,Wound Healing ,Wounds and Injuries ,Oncology and Carcinogenesis ,Dermatology & Venereal Diseases ,Clinical sciences - Abstract
In this study, we report that TIP39, a parathyroid hormone ligand family member that was recently identified to be expressed in the skin, can induce decorin expression and enhance wound repair. Topical treatment of mice with TIP39 accelerated wound repair, whereas TIP39-deficient mice had delayed repair that was associated with formation of abnormal collagen bundles. To study the potential mechanism responsible for the action of TIP39 in the dermis, fibroblasts were cultured in three-dimensional collagen gels, a process that results in enhanced decorin expression unless activated to differentiate to adipocytes, whereupon these cells reduce expression of several proteoglycans, including decorin. Small interfering RNA-mediated silencing of parathyroid hormone 2 receptor (PTH2R), the receptor for TIP39, suppressed the expression of extracellular matrix-related genes, including decorin, collagens, fibronectin, and matrix metalloproteases. Skin wounds in TIP39-/- mice had decreased decorin expression, and addition of TIP39 to cultured fibroblasts induced decorin and increased phosphorylation and nuclear translocation of CREB. Fibroblasts differentiated to adipocytes and treated with TIP39 also showed increased decorin and production of chondroitin sulfate. Furthermore, the skin of PTH2R-/- mice showed abnormal extracellular matrix structure, decreased decorin expression, and skin hardness. Thus, the TIP39-PTH2R system appears to be a previously unrecognized mechanism for regulation of extracellular matrix formation and wound repair.
- Published
- 2017
109. Neutral Competition for Drosophila Follicle and Cyst Stem Cell Niches Requires Vesicle Trafficking Genes
- Author
-
Cook, Matthew S, Cazin, Coralie, Amoyel, Marc, Yamamoto, Shinya, Bach, Erika, and Nystul, Todd
- Subjects
Biochemistry and Cell Biology ,Genetics ,Biological Sciences ,Stem Cell Research ,Stem Cell Research - Nonembryonic - Non-Human ,Regenerative Medicine ,Underpinning research ,1.1 Normal biological development and functioning ,Generic health relevance ,Animals ,Cell Differentiation ,Chromosomes ,Insect ,Drosophila ,Drosophila Proteins ,Female ,Male ,Ovarian Follicle ,Stem Cell Niche ,Testis ,Vesicular Transport Proteins ,X Chromosome ,cyst stem cell ,follicle stem cell ,neutral competition ,vesicle trafficking ,Developmental Biology ,Biochemistry and cell biology - Abstract
The process of selecting for cellular fitness through competition plays a critical role in both development and disease. The germarium, a structure at the tip of the ovariole of a Drosophila ovary, contains two follicle stem cells (FSCs) that undergo neutral competition for the stem cell niche. Using the FSCs as a model, we performed a genetic screen through a collection of 126 mutants in essential genes on the X chromosome to identify candidates that increase or decrease competition for the FSC niche. We identified ∼55 and 6% of the mutations screened as putative FSC hypo- or hyper-competitors, respectively. We found that a large majority of mutations in vesicle trafficking genes (11 out of the 13 in the collection of mutants) are candidate hypo-competition alleles, and we confirmed the hypo-competition phenotype for four of these alleles. We also show that Sec16 and another COPII vesicle trafficking component, Sar1, are required for follicle cell differentiation. Lastly, we demonstrate that, although some components of vesicle trafficking are also required for neutral competition in the cyst stem cells of the testis, there are important tissue-specific differences. Our results demonstrate a critical role for vesicle trafficking in stem cell niche competition and differentiation, and we identify a number of putative candidates for further exploration.
- Published
- 2017
110. Autophagy-Dependent Shuttling of TBC1D5 Controls Plasma Membrane Translocation of GLUT1 and Glucose Uptake.
- Author
-
Roy, Srirupa, Leidal, Andrew M, Ye, Jordan, Ronen, Sabrina M, and Debnath, Jayanta
- Subjects
Cell Membrane ,Endosomes ,Lysosomes ,Fibroblasts ,Animals ,Humans ,Mice ,Glucose ,GTPase-Activating Proteins ,Microtubule-Associated Proteins ,Vesicular Transport Proteins ,Transfection ,Signal Transduction ,RNA Interference ,Protein Transport ,Glycolysis ,Kinetics ,Autophagy ,Female ,Glucose Transporter Type 1 ,Stress ,Physiological ,HEK293 Cells ,Autophagy-Related Protein 5 ,Autophagy-Related Protein 7 ,Autophagosomes ,GLUT1 ,autophagy ,glycolysis ,retromer ,Generic health relevance ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
Autophagy traditionally sustains metabolism in stressed cells by promoting intracellular catabolism and nutrient recycling. Here, we demonstrate that in response to stresses requiring increased glycolytic demand, the core autophagy machinery also facilitates glucose uptake and glycolytic flux by promoting cell surface expression of the glucose transporter GLUT1/Slc2a1. During metabolic stress, LC3+ autophagic compartments bind and sequester the RabGAP protein TBC1D5 away from its inhibitory interactions with the retromer complex, thereby enabling retromer recruitment to endosome membranes and GLUT1 plasma membrane translocation. In contrast, TBC1D5 inhibitory interactions with the retromer are maintained in autophagy-deficient cells, leading to GLUT1 mis-sorting into endolysosomal compartments. Furthermore, TBC1D5 depletion in autophagy-deficient cells rescues retromer recruitment to endosomal membranes and GLUT1 surface recycling. Hence, TBC1D5 shuttling to autophagosomes during metabolic stress facilitates retromer-dependent GLUT1 trafficking. Overall, our results illuminate key interconnections between the autophagy and endosomal pathways dictating GLUT1 trafficking and extracellular nutrient uptake.
- Published
- 2017
111. Loss of WDFY3 ameliorates severity of serum transfer-induced arthritis independently of autophagy
- Author
-
Wu, Dennis J and Adamopoulos, Iannis E
- Subjects
Biomedical and Clinical Sciences ,Immunology ,Arthritis ,Autoimmune Disease ,2.1 Biological and endogenous factors ,Aetiology ,Adaptor Proteins ,Signal Transducing ,Animals ,Arthritis ,Experimental ,Autophagosomes ,Autophagy ,Autophagy-Related Proteins ,Cells ,Cultured ,Joint Diseases ,Macrophages ,Mice ,Mice ,Inbred C57BL ,Mice ,Knockout ,Osteoclasts ,Vesicular Transport Proteins ,ALFY ,Autophagy-linked FYVE containing protein ,Musculoskeletal diseases ,Osteoclast ,WDFY3 - Abstract
WDFY3 is a master regulator of selective autophagy that we recently showed to interact with TRAF6 and augment RANKL-induced osteoclastogenesis in vitro and in vivo via the NF-κB pathway. Since the NF-κB pathway plays a major role in inflammation herein, we investigate the role of WDFY3 in an arthritis animal model. Our data show that WDFY3 conditional knockout mice (Wdfy3loxP/loxP-LysM-Cre+) were protected in the K/BxN serum transfer-induced arthritis animal model. These effects were independent of alterations in starvation-induced autophagy as evidenced by Western blot analysis of the autophagy marker LC3, autophagosome formation in osteoclast precursors and lysosome formation in osteoclasts derived from WDFY3-cKO mice compared to controls. Moreover, we demonstrate by immunofluorescence and co-immunoprecipitation that WDFY3 interacts with SQSTM1 in macrophages and osteoclasts. Collectively, our data suggest that loss of WDFY3 in myeloid cells leads to reduced severity of inflammatory arthritis independently of WDFY3 function in starvation-induced autophagy.
- Published
- 2017
112. Acute acinar pancreatitis blocks vesicle-associated membrane protein 8 (VAMP8)-dependent secretion, resulting in intracellular trypsin accumulation
- Author
-
Messenger, Scott W, Jones, Elaina K, Holthaus, Conner L, Thomas, Diana DH, Cooley, Michelle M, Byrne, Jennifer A, Mareninova, Olga A, Gukovskaya, Anna S, and Groblewski, Guy E
- Subjects
Biochemistry and Cell Biology ,Medical Physiology ,Biomedical and Clinical Sciences ,Biological Sciences ,Digestive Diseases ,2.1 Biological and endogenous factors ,Aetiology ,Oral and gastrointestinal ,Animals ,Endosomes ,Female ,Kinetics ,Male ,Mice ,Mice ,Inbred C57BL ,Pancreas ,Pancreatitis ,R-SNARE Proteins ,Rats ,Rats ,Sprague-Dawley ,Trypsin ,Trypsinogen ,Vesicular Transport Proteins ,rab5 GTP-Binding Proteins ,SNARE proteins ,autophagy ,endosome ,pancreas ,trypsin ,trypsinogen ,Chemical Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
Zymogen secretory granules in pancreatic acinar cells express two vesicle-associated membrane proteins (VAMP), VAMP2 and -8, each controlling 50% of stimulated secretion. Analysis of secretion kinetics identified a first phase (0-2 min) mediated by VAMP2 and second (2-10 min) and third phases (10-30 min) mediated by VAMP8. Induction of acinar pancreatitis by supramaximal cholecystokinin (CCK-8) stimulation inhibits VAMP8-mediated mid- and late-phase but not VAMP2-mediated early-phase secretion. Elevation of cAMP during supramaximal CCK-8 mitigates third-phase secretory inhibition and acinar damage caused by the accumulation of prematurely activated trypsin. VAMP8-/- acini are resistant to secretory inhibition by supramaximal CCK-8, and despite a 4.5-fold increase in total cellular trypsinogen levels, are fully protected from intracellular trypsin accumulation and acinar damage. VAMP8-mediated secretion is dependent on expression of the early endosomal proteins Rab5, D52, and EEA1. Supramaximal CCK-8 (60 min) caused a 60% reduction in the expression of D52 followed by Rab5 and EEA1 in isolated acini and in in vivo The loss of D52 occurred as a consequence of its entry into autophagic vacuoles and was blocked by lysosomal cathepsin B and L inhibition. Accordingly, adenoviral overexpression of Rab5 or D52 enhanced secretion in response to supramaximal CCK-8 and prevented accumulation of activated trypsin. These data support that acute inhibition of VAMP8-mediated secretion during pancreatitis triggers intracellular trypsin accumulation and loss of the early endosomal compartment. Maintaining anterograde endosomal trafficking during pancreatitis maintains VAMP8-dependent secretion, thereby preventing accumulation of activated trypsin.
- Published
- 2017
113. LYVE1 Marks the Divergence of Yolk Sac Definitive Hemogenic Endothelium from the Primitive Erythroid Lineage.
- Author
-
Lee, Lydia K, Ghorbanian, Yasamine, Wang, Wenyuan, Wang, Yanling, Kim, Yeon Joo, Weissman, Irving L, Inlay, Matthew A, and Mikkola, Hanna KA
- Subjects
Liver ,Yolk Sac ,Hematopoietic Stem Cells ,Erythroid Cells ,Fetus ,Animals ,Mice ,Inbred C57BL ,Integrases ,Vesicular Transport Proteins ,Erythropoiesis ,Gene Deletion ,Cell Lineage ,Aging ,Pregnancy ,Time Factors ,Female ,Hemangioblasts ,LYVE1 ,definitive hematopoiesis ,fetal liver ,hemogenic endothelium ,lineage tracing ,primitive hematopoiesis ,yolk sac ,Stem Cell Research ,Regenerative Medicine ,Hematology ,Stem Cell Research - Nonembryonic - Non-Human ,Transplantation ,Biochemistry and Cell Biology ,Medical Physiology - Abstract
The contribution of the different waves and sites of developmental hematopoiesis to fetal and adult blood production remains unclear. Here, we identify lymphatic vessel endothelial hyaluronan receptor-1 (LYVE1) as a marker of yolk sac (YS) endothelium and definitive hematopoietic stem and progenitor cells (HSPCs). Endothelium in mid-gestation YS and vitelline vessels, but not the dorsal aorta and placenta, were labeled by Lyve1-Cre. Most YS HSPCs and erythro-myeloid progenitors were Lyve1-Cre lineage traced, but primitive erythroid cells were not, suggesting that they represent distinct lineages. Fetal liver (FL) and adult HSPCs showed 35%-40% Lyve1-Cre marking. Analysis of circulation-deficient Ncx1-/- concepti identified the YS as a major source of Lyve1-Cre labeled HSPCs. FL proerythroblast marking was extensive at embryonic day (E) 11.5-13.5, but decreased to hematopoietic stem cell (HSC) levels by E16.5, suggesting that HSCs from multiple sources became responsible for erythropoiesis. Lyve1-Cre thus marks the divergence between YS primitive and definitive hematopoiesis and provides a tool for targeting YS definitive hematopoiesis and FL colonization.
- Published
- 2016
114. GIV/Girdin activates Gαi and inhibits Gαs via the same motif
- Author
-
Gupta, Vijay, Bhandari, Deepali, Leyme, Anthony, Aznar, Nicolas, Midde, Krishna K, Lo, I-Chung, Ear, Jason, Niesman, Ingrid, López-Sánchez, Inmaculada, Blanco-Canosa, Juan Bautista, von Zastrow, Mark, Garcia-Marcos, Mikel, Farquhar, Marilyn G, and Ghosh, Pradipta
- Subjects
1.1 Normal biological development and functioning ,Underpinning research ,Generic health relevance ,Amino Acid Motifs ,Amino Acid Sequence ,Cell Proliferation ,Chemotaxis ,Cyclic AMP ,Cyclic AMP Response Element-Binding Protein ,Cyclic AMP-Dependent Protein Kinases ,Cyclin-Dependent Kinase 5 ,Down-Regulation ,Endosomes ,Epidermal Growth Factor ,Extracellular Signal-Regulated MAP Kinases ,Fluorescence Resonance Energy Transfer ,GTP-Binding Protein alpha Subunits ,Gi-Go ,GTP-Binding Protein alpha Subunits ,Gs ,GTP-Binding Protein beta Subunits ,GTP-Binding Protein gamma Subunits ,Guanosine Triphosphate ,HeLa Cells ,Humans ,Microfilament Proteins ,Mutant Proteins ,Phosphorylation ,Protein Binding ,Protein Kinase C-theta ,Signal Transduction ,Structure-Activity Relationship ,Vesicular Transport Proteins ,heterotrimeric G proteins ,cAMP ,cancer invasion ,growth factor receptor tyrosine kinase ,guanine nucleotide dissociation inhibitor ,Hela Cells - Abstract
We previously showed that guanine nucleotide-binding (G) protein α subunit (Gα)-interacting vesicle-associated protein (GIV), a guanine-nucleotide exchange factor (GEF), transactivates Gα activity-inhibiting polypeptide 1 (Gαi) proteins in response to growth factors, such as EGF, using a short C-terminal motif. Subsequent work demonstrated that GIV also binds Gαs and that inactive Gαs promotes maturation of endosomes and shuts down mitogenic MAPK-ERK1/2 signals from endosomes. However, the mechanism and consequences of dual coupling of GIV to two G proteins, Gαi and Gαs, remained unknown. Here we report that GIV is a bifunctional modulator of G proteins; it serves as a guanine nucleotide dissociation inhibitor (GDI) for Gαs using the same motif that allows it to serve as a GEF for Gαi. Upon EGF stimulation, GIV modulates Gαi and Gαs sequentially: first, a key phosphomodification favors the assembly of GIV-Gαi complexes and activates GIV's GEF function; then a second phosphomodification terminates GIV's GEF function, triggers the assembly of GIV-Gαs complexes, and activates GIV's GDI function. By comparing WT and GIV mutants, we demonstrate that GIV inhibits Gαs activity in cells responding to EGF. Consequently, the cAMP→PKA→cAMP response element-binding protein signaling axis is inhibited, the transit time of EGF receptor through early endosomes are accelerated, mitogenic MAPK-ERK1/2 signals are rapidly terminated, and proliferation is suppressed. These insights define a paradigm in G-protein signaling in which a pleiotropically acting modulator uses the same motif both to activate and to inhibit G proteins. Our findings also illuminate how such modulation of two opposing Gα proteins integrates downstream signals and cellular responses.
- Published
- 2016
115. Autophagy-linked FYVE containing protein WDFY3 interacts with TRAF6 and modulates RANKL-induced osteoclastogenesis
- Author
-
Wu, Dennis J, Gu, Ran, Sarin, Ritu, Zavodovskaya, Regina, Chen, Chia-Pei, Christiansen, Blaine A, Zarbalis, Konstantinos S, and Adamopoulos, Iannis E
- Subjects
Biomedical and Clinical Sciences ,Immunology ,Autoimmune Disease ,Biotechnology ,Genetics ,Arthritis ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Musculoskeletal ,Inflammatory and immune system ,Adaptor Proteins ,Signal Transducing ,Animals ,Autophagy ,Autophagy-Related Proteins ,Blotting ,Western ,Bone Resorption ,Cathepsin K ,Cell Differentiation ,Cells ,Cultured ,Femur ,Gene Transfer Techniques ,Giant Cells ,Macrophages ,Matrix Metalloproteinase 9 ,Mice ,Mice ,Inbred C57BL ,Mice ,Knockout ,NF-kappa B ,Osteoblasts ,Osteoclasts ,Osteogenesis ,Primary Cell Culture ,RANK Ligand ,Signal Transduction ,TNF Receptor-Associated Factor 6 ,Tartrate-Resistant Acid Phosphatase ,Up-Regulation ,Vesicular Transport Proteins ,X-Ray Microtomography ,Autophagy-linked FYVE containing protein ,WDFY3 ,TRAF6 ,Osteoclast ,Musculoskeletal diseases - Abstract
Recently, autophagy-related proteins were shown to regulate osteoclast mediated bone resorption, a critical process in autoimmune diseases such as rheumatoid arthritis. However, the role of autophagy-linked FYVE containing protein, WDFY3, in osteoclast biology remains elusive. WDFY3 is a master regulator in selective autophagy for clearing ubiquitinated protein aggregates and has been linked with rheumatoid arthritis. Herein, we used a series of WDFY3 transgenic mice (Wdfy3(lacZ) and Wdfy3(loxP)) to investigate the function of WDFY3 in osteoclast development and function. Our data demonstrate that WDFY3 is highly expressed at the growth plate of neonatal mice and is expressed in osteoclasts in vitro cultures. Osteoclasts derived from WDFY3 conditional knockout mice (Wdfy3(loxP/loxP)-LysM-Cre(+)) demonstrated increased osteoclast differentiation as evidenced by higher number and enlarged size of TRAP(+) multinucleated cells. Western blot analysis also revealed up-regulation of TRAF6 and an increase in RANKL-induced NF-κB signaling in WDFY3-deficient bone marrow-derived macrophages compared to wild type cultures. Consistent with these observations WDFY3-deficient cells also demonstrated an increase in osteoclast-related genes Ctsk, Acp5, Mmp9 and an increase of dentine resorption in in vitro assays. Importantly, in vivo RANKL gene transfer exacerbated bone loss in WDFY3 conditional knockout mice, as evidenced by elevated serum TRAP, CTX-I and micro-CT analysis of distal femurs compared to wild type littermates. Taken together, our data highlight a novel role for WDFY3 in osteoclast development and function, which can be exploited for the treatment of musculoskeletal diseases.
- Published
- 2016
116. Girdin (GIV) Expression as a Prognostic Marker of Recurrence in Mismatch Repair–Proficient Stage II Colon Cancer
- Author
-
Ghosh, Pradipta, Tie, Jeanne, Muranyi, Andrea, Singh, Shalini, Brunhoeber, Patrick, Leith, Katherine, Bowermaster, Rebecca, Liao, Zhiming, Zhu, Yifei, LaFleur, Bonnie, Tran, Ben, Desai, Jayesh, Jones, Ian, Croxford, Matthew, Jover, Rodrigo, Goel, Ajay, Waring, Paul, Hu, Song, Teichgraber, Volker, Rohr, Ulrich-Peter, Ridder, Ruediger, Shanmugam, Kandavel, and Gibbs, Peter
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Colo-Rectal Cancer ,Cancer ,Digestive Diseases ,Detection ,screening and diagnosis ,4.2 Evaluation of markers and technologies ,Aged ,Biomarkers ,Tumor ,Chemotherapy ,Adjuvant ,Colonic Neoplasms ,DNA Mismatch Repair ,Disease-Free Survival ,Female ,Humans ,Male ,Microfilament Proteins ,Neoplasm Invasiveness ,Neoplasm Recurrence ,Local ,Neoplasm Staging ,Prognosis ,Vesicular Transport Proteins ,Oncology & Carcinogenesis ,Clinical sciences ,Oncology and carcinogenesis - Abstract
PurposePrognostic markers that identify patients with stage II colon cancers who are at the risk of recurrence are essential to personalize therapy. We evaluated the potential of GIV/Girdin as a predictor of recurrence risk in such patients.Experimental designExpression of full-length GIV was evaluated by IHC using a newly developed mAb together with a mismatch repair (MMR)-specific antibody panel in three stage II colon cancer patient cohorts, that is, a training (n = 192), test (n = 317), and validation (n = 181) cohort, with clinical follow-up data. Recurrence risk stratification models were established in the training cohort of T3, proficient MMR (pMMR) patients without chemotherapy and subsequently validated.ResultsFor T3 pMMR tumors, GIV expression and the presence of lymphovascular invasion (LVI) were the only factors predicting recurrence in both training (GIV: HR, 2.78, P = 0.013; LVI: HR, 2.54, P = 0.025) and combined test and validation (pooled) cohorts (GIV: HR, 1.85, P = 0.019; LVI: HR, 2.52, P = 0.0004). A risk model based on GIV expression and LVI status classified patients into high- or low-risk groups; 3-year recurrence-free survival was significantly lower in the high-risk versus low-risk group across all cohorts [Training: 52.3% vs. 84.8%; HR, 3.74, 95% confidence interval (CI), 1.50-9.32; Test: 85.9% vs. 97.9%, HR, 7.83, 95% CI, 1.03-59.54; validation: 59.4% vs. 84.4%, HR, 3.71, 95% CI, 1.24-11.12].ConclusionsGIV expression status predicts recurrence risk in patients with T3 pMMR stage II colon cancer. A risk model combining GIV expression and LVI status information further enhances prediction of recurrence. Further validation studies are warranted before GIV status can be routinely included in patient management algorithms. Clin Cancer Res; 22(14); 3488-98. ©2016 AACR.
- Published
- 2016
117. Distinct stages in the recognition, sorting, and packaging of proTGFα into COPII-coated transport vesicles
- Author
-
Zhang, Pengcheng and Schekman, Randy
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Rare Diseases ,Brain Disorders ,Underpinning research ,1.1 Normal biological development and functioning ,Generic health relevance ,Animals ,Biological Transport ,COP-Coated Vesicles ,Carrier Proteins ,Cell Line ,Cytosol ,Endoplasmic Reticulum ,Humans ,Membrane Proteins ,Protein Precursors ,Protein Transport ,Transforming Growth Factor alpha ,Transport Vesicles ,Vesicular Transport Proteins ,Medical and Health Sciences ,Developmental Biology ,Biochemistry and cell biology - Abstract
In addition to its role in forming vesicles from the endoplasmic reticulum (ER), the coat protein complex II (COPII) is also responsible for selecting specific cargo proteins to be packaged into COPII transport vesicles. Comparison of COPII vesicle formation in mammalian systems and in yeast suggested that the former uses more elaborate mechanisms for cargo recognition, presumably to cope with a significantly expanded repertoire of cargo that transits the secretory pathway. Using proTGFα, the transmembrane precursor of transforming growth factor α (TGFα), as a model cargo protein, we demonstrate in cell-free assays that at least one auxiliary cytosolic factor is specifically required for the efficient packaging of proTGFα into COPII vesicles. Using a knockout HeLa cell line generated by CRISPR/Cas9, we provide functional evidence showing that a transmembrane protein, Cornichon-1 (CNIH), acts as a cargo receptor of proTGFα. We show that both CNIH and the auxiliary cytosolic factor(s) are required for efficient recruitment of proTGFα to the COPII coat in vitro. Moreover, we provide evidence that the recruitment of cargo protein by the COPII coat precedes and may be distinct from subsequent cargo packaging into COPII vesicles.
- Published
- 2016
118. Organelle Size Scaling of the Budding Yeast Vacuole by Relative Growth and Inheritance
- Author
-
Chan, Yee-Hung M, Reyes, Lorena, Sohail, Saba M, Tran, Nancy K, and Marshall, Wallace F
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Generic health relevance ,Gene Expression Regulation ,Fungal ,Organelles ,Saccharomyces cerevisiae ,Saccharomyces cerevisiae Proteins ,Vacuoles ,Vesicular Transport Proteins ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Psychology - Abstract
It has long been noted that larger animals have larger organs compared to smaller animals of the same species, a phenomenon termed scaling [1]. Julian Huxley proposed an appealingly simple model of "relative growth"-in which an organ and the whole body grow with their own intrinsic rates [2]-that was invoked to explain scaling in organs from fiddler crab claws to human brains. Because organ size is regulated by complex, unpredictable pathways [3], it remains unclear whether scaling requires feedback mechanisms to regulate organ growth in response to organ or body size. The molecular pathways governing organelle biogenesis are simpler than organogenesis, and therefore organelle size scaling in the cell provides a more tractable case for testing Huxley's model. We ask the question: is it possible for organelle size scaling to arise if organelle growth is independent of organelle or cell size? Using the yeast vacuole as a model, we tested whether mutants defective in vacuole inheritance, vac8Δ and vac17Δ, tune vacuole biogenesis in response to perturbations in vacuole size. In vac8Δ/vac17Δ, vacuole scaling increases with the replicative age of the cell. Furthermore, vac8Δ/vac17Δ cells continued generating vacuole at roughly constant rates even when they had significantly larger vacuoles compared to wild-type. With support from computational modeling, these results suggest there is no feedback between vacuole biogenesis rates and vacuole or cell size. Rather, size scaling is determined by the relative growth rates of the vacuole and the cell, thus representing a cellular version of Huxley's model.
- Published
- 2016
119. Distinct Lysosomal Network Protein Profiles in Parkinsonian Syndrome Cerebrospinal Fluid
- Author
-
Boman, Andrea, Svensson, Samuel, Boxer, Adam, Rojas, Julio C, Seeley, William W, Karydas, Anna, Miller, Bruce, Kågedal, Katarina, and Svenningsson, Per
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Brain Disorders ,Orphan Drug ,Clinical Research ,Rare Diseases ,Acquired Cognitive Impairment ,Dementia ,Parkinson's Disease ,Neurodegenerative ,Aging ,Aetiology ,2.1 Biological and endogenous factors ,4.2 Evaluation of markers and technologies ,Detection ,screening and diagnosis ,Neurological ,Adult ,Aged ,Aged ,80 and over ,Biomarkers ,Female ,Humans ,Lysosomal-Associated Membrane Protein 2 ,Lysosome-Associated Membrane Glycoproteins ,Lysosomes ,Male ,Microtubule-Associated Proteins ,Middle Aged ,Parkinsonian Disorders ,Supranuclear Palsy ,Progressive ,Tauopathies ,Vesicular Transport Proteins ,CSF ,parkinson's disease ,corticobasal degeneration ,progressive supranuclear palsy ,parkinson’s disease ,Biochemistry and Cell Biology - Abstract
BackgroundClinical diagnosis of parkinsonian syndromes like Parkinson's disease (PD), corticobasal degeneration (CBD) and progressive supranuclear palsy (PSP) is hampered by overlapping symptomatology and lack of diagnostic biomarkers, and definitive diagnosis is only possible post-mortem.ObjectiveSince impaired protein degradation plays an important role in many neurodegenerative disorders, we hypothesized that profiles of select lysosomal network proteins in cerebrospinal fluid could be differentially expressed in these parkinsonian syndromes.MethodsCerebrospinal fluid samples were collected from PD patients (n = 18), clinically diagnosed 4-repeat tauopathy patients; corticobasal syndrome (CBS) (n = 3) and PSP (n = 8); and pathologically diagnosed PSP (n = 8) and CBD patients (n = 7). Each patient set was compared to its appropriate control group consisting of age and gender matched individuals. Select lysosomal network protein levels were detected via Western blotting. Factor analysis was used to test the diagnostic sensitivity, specificity and accuracy of the select lysosomal network protein expression profiles.ResultsPD, CBD and PSP were markedly different in their cerebrospinal fluid lysosomal network protein profiles. Lysosomal-associated membrane proteins 1 and 2 were significantly decreased in PD; early endosomal antigen 1 was decreased and lysozyme increased in PSP; and lysosomal-associated membrane proteins 1 and 2, microtubule-associated protein 1 light chain 3 and lysozyme were increased in CBD. A panel of lysosomal-associated membrane protein 2, lysozyme and microtubule-associated protein 1 light chain discriminated between controls, PD and 4-repeat tauopathies.ConclusionsThis study offers proof of concept that select lysosomal network proteins are differentially expressed in cerebrospinal fluid of Parkinson's disease, corticobasal syndrome and progressive supranuclear palsy. Lysosomal network protein analysis could be further developed as a diagnostic fluid biomarker in parkinsonian syndromes.
- Published
- 2016
120. Heterotrimeric G protein signaling via GIV/Girdin: Breaking the rules of engagement, space, and time
- Author
-
Aznar, Nicolas, Kalogriopoulos, Nicholas, Midde, Krishna K, and Ghosh, Pradipta
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,1.1 Normal biological development and functioning ,Underpinning research ,Generic health relevance ,Cardiovascular Diseases ,Cell Membrane ,Diabetes Mellitus ,Gene Expression Regulation ,Gene Regulatory Networks ,Humans ,Intracellular Membranes ,Microfilament Proteins ,Models ,Molecular ,Molecular Targeted Therapy ,Neoplasms ,Protein Interaction Mapping ,Receptors ,G-Protein-Coupled ,Signal Transduction ,Time Factors ,Vesicular Transport Proteins ,autophagy ,cdk5: Girdin ,Golgi ,growth factor receptor tyrosine kinases ,heterotrimeric G proteins ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Developmental Biology ,Biological sciences - Abstract
Canonical signal transduction via heterotrimeric G proteins is spatially and temporally restricted, that is, triggered exclusively at the plasma membrane (PM), only by agonist activation of G protein-coupled receptors (GPCRs) via a process that completes within a few hundred milliseconds. Recently, a rapidly emerging paradigm has revealed a non-canonical pathway for activation of heterotrimeric G proteins by the non-receptor guanidine-nucleotide exchange factor (GEF), GIV/Girdin. This pathway has distinctive temporal and spatial features and an unusual profile of receptor engagement: diverse classes of receptors, not just GPCRs can engage with GIV to trigger such activation. Such activation is spatially and temporally unrestricted, that is, can occur both at the PM and on internal membranes discontinuous with the PM, and can continue for prolonged periods of time. Here, we provide the most complete up-to-date review of the molecular mechanisms that govern the unique spatiotemporal aspects of non-canonical G protein activation by GIV and the relevance of this new paradigm in health and disease.
- Published
- 2016
121. Ang-2/VEGF bispecific antibody reprograms macrophages and resident microglia to anti-tumor phenotype and prolongs glioblastoma survival.
- Author
-
Kloepper, Jonas, Riedemann, Lars, Amoozgar, Zohreh, Seano, Giorgio, Susek, Katharina, Yu, Veronica, Dalvie, Nisha, Amelung, Robin L, Datta, Meenal, Song, Jonathan W, Askoxylakis, Vasileios, Taylor, Jennie W, Lu-Emerson, Christine, Batista, Ana, Kirkpatrick, Nathaniel D, Jung, Keehoon, Snuderl, Matija, Muzikansky, Alona, Stubenrauch, Kay G, Krieter, Oliver, Wakimoto, Hiroaki, Xu, Lei, Munn, Lance L, Duda, Dan G, Fukumura, Dai, Batchelor, Tracy T, and Jain, Rakesh K
- Subjects
Cell Line ,Tumor ,Macrophages ,Animals ,Humans ,Mice ,Glioblastoma ,Neoplasms ,Experimental ,Ribonuclease ,Pancreatic ,Vascular Endothelial Growth Factor A ,Vesicular Transport Proteins ,Neoplasm Proteins ,Antineoplastic Agents ,Antibodies ,Bispecific ,Antibodies ,Neoplasm ,Xenograft Model Antitumor Assays ,anti-angiogenic therapy ,anti-tumor immunity ,macrophage polarization ,microglia reprogramming ,tumor microenvironment ,Cancer ,Biotechnology ,Brain Disorders ,Brain Cancer ,Rare Diseases ,Neurosciences ,2.1 Biological and endogenous factors ,Aetiology - Abstract
Inhibition of the vascular endothelial growth factor (VEGF) pathway has failed to improve overall survival of patients with glioblastoma (GBM). We previously showed that angiopoietin-2 (Ang-2) overexpression compromised the benefit from anti-VEGF therapy in a preclinical GBM model. Here we investigated whether dual Ang-2/VEGF inhibition could overcome resistance to anti-VEGF treatment. We treated mice bearing orthotopic syngeneic (Gl261) GBMs or human (MGG8) GBM xenografts with antibodies inhibiting VEGF (B20), or Ang-2/VEGF (CrossMab, A2V). We examined the effects of treatment on the tumor vasculature, immune cell populations, tumor growth, and survival in both the Gl261 and MGG8 tumor models. We found that in the Gl261 model, which displays a highly abnormal tumor vasculature, A2V decreased vessel density, delayed tumor growth, and prolonged survival compared with B20. In the MGG8 model, which displays a low degree of vessel abnormality, A2V induced no significant changes in the tumor vasculature but still prolonged survival. In both the Gl261 and MGG8 models A2V reprogrammed protumor M2 macrophages toward the antitumor M1 phenotype. Our findings indicate that A2V may prolong survival in mice with GBM by reprogramming the tumor immune microenvironment and delaying tumor growth.
- Published
- 2016
122. Prognostic Impact of Modulators of G proteins in Circulating Tumor Cells from Patients with Metastatic Colorectal Cancer.
- Author
-
Barbazan, Jorge, Dunkel, Ying, Li, Hongying, Nitsche, Ulrich, Janssen, Klaus-Peter, Messer, Karen, and Ghosh, Pradipta
- Subjects
Humans ,Colorectal Neoplasms ,Neoplasm Metastasis ,Microfilament Proteins ,GTP-Binding Protein alpha Subunits ,Intracellular Signaling Peptides and Proteins ,Guanine Nucleotide Exchange Factors ,Calcium-Binding Proteins ,DNA-Binding Proteins ,Vesicular Transport Proteins ,Nerve Tissue Proteins ,Transcription Factors ,Disease-Free Survival ,Gene Expression ,Protein Structure ,Tertiary ,Neoplastic Cells ,Circulating ,Biomarkers ,Tumor ,S100 Calcium-Binding Protein A4 ,Nucleobindins ,Biomarkers ,Tumor ,Neoplastic Cells ,Circulating ,Protein Structure ,Tertiary - Abstract
The consequence of a loss of balance between G-protein activation and deactivation in cancers has been interrogated by studying infrequently occurring mutants of trimeric G-protein α-subunits and GPCRs. Prior studies on members of a newly identified family of non-receptor guanine nucleotide exchange factors (GEFs), GIV/Girdin, Daple, NUCB1 and NUCB2 have revealed that GPCR-independent hyperactivation of trimeric G proteins can fuel metastatic progression in a variety of cancers. Here we report that elevated expression of each GEF in circulating tumor cells (CTCs) isolated from the peripheral circulation of patients with metastatic colorectal cancer is associated with a shorter progression-free survival (PFS). The GEFs were stronger prognostic markers than two other markers of cancer progression, S100A4 and MACC1, and clustering of all GEFs together improved the prognostic accuracy of the individual family members; PFS was significantly lower in the high-GEFs versus the low-GEFs groups [H.R = 5, 20 (95% CI; 2,15-12,57)]. Because nucleotide exchange is the rate-limiting step in cyclical activation of G-proteins, the poor prognosis conferred by these GEFs in CTCs implies that hyperactivation of G-protein signaling by these GEFs is an important event during metastatic progression, and may be more frequently encountered than mutations in G-proteins and/or GPCRs.
- Published
- 2016
123. The casein kinases Yck1p and Yck2p act in the secretory pathway, in part, by regulating the Rab exchange factor Sec2p
- Author
-
Stalder, Danièle and Novick, Peter J
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Underpinning research ,1.1 Normal biological development and functioning ,Generic health relevance ,Casein Kinase I ,Glucan Endo-1 ,3-beta-D-Glucosidase ,Golgi Apparatus ,Guanine Nucleotide Exchange Factors ,Mutation ,Phosphatidylinositol Phosphates ,Phosphorylation ,Protein Binding ,Protein Transport ,Saccharomyces cerevisiae ,Saccharomyces cerevisiae Proteins ,Secretory Pathway ,Secretory Vesicles ,Vesicular Transport Proteins ,rab GTP-Binding Proteins ,Medical and Health Sciences ,Developmental Biology ,Biochemistry and cell biology - Abstract
Sec2p is a guanine nucleotide exchange factor that activates Sec4p, the final Rab GTPase of the yeast secretory pathway. Sec2p is recruited to secretory vesicles by the upstream Rab Ypt32p acting in concert with phosphatidylinositol-4-phosphate (PI(4)P). Sec2p also binds to the Sec4p effector Sec15p, yet Ypt32p and Sec15p compete against each other for binding to Sec2p. We report here that the redundant casein kinases Yck1p and Yck2p phosphorylate sites within the Ypt32p/Sec15p binding region and in doing so promote binding to Sec15p and inhibit binding to Ypt32p. We show that Yck2p binds to the autoinhibitory domain of Sec2p, adjacent to the PI(4)P binding site, and that addition of PI(4)P inhibits Sec2p phosphorylation by Yck2p. Loss of Yck1p and Yck2p function leads to accumulation of an intracellular pool of the secreted glucanase Bgl2p, as well as to accumulation of Golgi-related structures in the cytoplasm. We propose that Sec2p is phosphorylated after it has been recruited to secretory vesicles and the level of PI(4)P has been reduced. This promotes Sec2p function by stimulating its interaction with Sec15p. Finally, Sec2p is dephosphorylated very late in the exocytic reaction to facilitate recycling.
- Published
- 2016
124. Subunit connectivity, assembly determinants and architecture of the yeast exocyst complex
- Author
-
Heider, Margaret R, Gu, Mingyu, Duffy, Caroline M, Mirza, Anne M, Marcotte, Laura L, Walls, Alexandra C, Farrall, Nicholas, Hakhverdyan, Zhanna, Field, Mark C, Rout, Michael P, Frost, Adam, and Munson, Mary
- Subjects
Genetics ,Bioengineering ,Exocytosis ,Macromolecular Substances ,Microscopy ,Electron ,Transmission ,Protein Binding ,Protein Stability ,Protein Structure ,Quaternary ,Saccharomyces cerevisiae ,Vesicular Transport Proteins ,Chemical Sciences ,Biological Sciences ,Medical and Health Sciences ,Biophysics ,Developmental Biology - Abstract
The exocyst is a hetero-octameric complex that has been proposed to serve as the tethering complex for exocytosis, although it remains poorly understood at the molecular level. Here, we purified endogenous exocyst complexes from Saccharomyces cerevisiae and showed that they are stable and consist of all eight subunits with equal stoichiometry. Using a combination of biochemical and auxin induced-degradation experiments in yeast, we mapped the subunit connectivity, identified two stable four-subunit modules within the octamer and demonstrated that several known exocyst-binding partners are not necessary for exocyst assembly and stability. Furthermore, we visualized the structure of the yeast complex by using negative-stain electron microscopy; our results indicate that the exocyst exists predominantly as a stable, octameric complex with an elongated architecture that suggests that the subunits are contiguous helical bundles packed together into a bundle of long rods.
- Published
- 2016
125. AMP-activated protein kinase fortifies epithelial tight junctions during energetic stress via its effector GIV/Girdin
- Author
-
Aznar, Nicolas, Patel, Arjun, Rohena, Cristina C, Dunkel, Ying, Joosen, Linda P, Taupin, Vanessa, Kufareva, Irina, Farquhar, Marilyn G, and Ghosh, Pradipta
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Cancer ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Generic health relevance ,AMP-Activated Protein Kinases ,Animals ,Cell Line ,Humans ,Microfilament Proteins ,Phosphorylation ,Protein Processing ,Post-Translational ,Tight Junctions ,Vesicular Transport Proteins ,Colon cancer ,Metformin ,AMPK ,cancer biology ,ccdc88a/GIRDIN ,cell biology ,energetic stress ,epithelial cell polarity ,human ,tight junctions ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
Loss of epithelial polarity impacts organ development and function; it is also oncogenic. AMPK, a key sensor of metabolic stress stabilizes cell-cell junctions and maintains epithelial polarity; its activation by Metformin protects the epithelial barrier against stress and suppresses tumorigenesis. How AMPK protects the epithelium remains unknown. Here, we identify GIV/Girdin as a novel effector of AMPK, whose phosphorylation at a single site is both necessary and sufficient for strengthening mammalian epithelial tight junctions and preserving cell polarity and barrier function in the face of energetic stress. Expression of an oncogenic mutant of GIV (cataloged in TCGA) that cannot be phosphorylated by AMPK increased anchorage-independent growth of tumor cells and helped these cells to evade the tumor-suppressive action of Metformin. This work defines a fundamental homeostatic mechanism by which the AMPK-GIV axis reinforces cell junctions against stress-induced collapse and also provides mechanistic insight into the tumor-suppressive action of Metformin.
- Published
- 2016
126. GIV/girdin binds exocyst subunit-Exo70 and regulates exocytosis of GLUT4 storage vesicles
- Author
-
Lopez-Sanchez, Inmaculada, S., Gary, Pedram, Shabnam, Kalogriopoulos, Nicholas, and Ghosh, Pradipta
- Subjects
Genetics ,Diabetes ,Metabolic and endocrine ,Animals ,Cell Line ,Exocytosis ,Glucose Transporter Type 4 ,Guanine Nucleotide Exchange Factors ,HeLa Cells ,Humans ,Insulin ,Microfilament Proteins ,Protein Binding ,Protein Transport ,Rats ,Signal Transduction ,Vesicular Transport Proteins ,Girdin/GIV ,PI3-Kinase ,Akt ,Glucose transporter/GLUT4 ,Exo-70 ,Exocyst complex ,Hela Cells ,Exo−70 ,PI3−Kinase ,Medicinal and Biomolecular Chemistry ,Biochemistry and Cell Biology ,Medical Biochemistry and Metabolomics ,Biochemistry & Molecular Biology - Abstract
Insulin resistance (IR) is a metabolic disorder characterized by impaired glucose uptake in response to insulin. The current paradigm for insulin signaling centers upon the insulin receptor (InsR) and its substrate IRS1; the latter is believed to be the chief conduit for post-receptor signaling. We recently demonstrated that GIV, a Guanidine Exchange Factor (GEF) for the trimeric G protein, Gαi, is a major hierarchical conduit for the metabolic insulin response. By virtue of its ability to directly bind the InsR, IRS1 and PI3K, GIV enhances the InsR-IRS1-Akt-AS160 (RabGAP) signaling cascade and cellular glucose uptake via its GEF function. Phosphoinhibition of GIV-GEF by the fatty-acid/PKCθ pathway inhibits the cascade and impairs glucose uptake. Here we show that GIV directly and constitutively binds the exocyst complex subunit Exo-70 and also associates with GLUT4-storage vesicles (GSVs) exclusively upon insulin stimulation. Without GIV or its GEF function, membrane association of Exo-70 as well as exocytosis of GSVs in response to insulin are impaired. Thus, GIV is an essential component within the insulin signaling cascade that couples upstream signal transducers within the InsR and G-Protein signaling cascade to downstream vesicular trafficking events within the exocytic pathway. These findings suggest a role of GIV in coordinating key signaling and trafficking events of metabolic insulin response.
- Published
- 2015
127. Novel VPS13B Mutations in Three Large Pakistani Cohen Syndrome Families Suggests a Baloch Variant with Autistic-Like Features
- Author
-
Rafiq, Muhammad Arshad, Leblond, Claire S, Saqib, Muhammad Arif Nadeem, Vincent, Akshita K, Ambalavanan, Amirthagowri, Khan, Falak Sher, Ayaz, Muhammad, Shaheen, Naseema, Spiegelman, Dan, Ali, Ghazanfar, Amin-ud-din, Muhammad, Laurent, Sandra, Mahmood, Huda, Christian, Mehtab, Ali, Nadir, Fennell, Alanna, Nanjiani, Zohair, Egger, Gerald, Caron, Chantal, Waqas, Ahmed, Ayub, Muhammad, Rasheed, Saima, Forgeot d’Arc, Baudouin, Johnson, Amelie, So, Joyce, Brohi, Muhammad Qasim, Mottron, Laurent, Ansar, Muhammad, Vincent, John B, and Xiong, Lan
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Mental Health ,Pediatric ,Rare Diseases ,Clinical Research ,Intellectual and Developmental Disabilities (IDD) ,Autism ,Brain Disorders ,Human Genome ,Aetiology ,2.1 Biological and endogenous factors ,Congenital ,Mental health ,Abnormalities ,Multiple ,Autistic Disorder ,Base Sequence ,Developmental Disabilities ,Female ,Fingers ,Genes ,Recessive ,Genotype ,Haplotypes ,Homozygote ,Humans ,Intellectual Disability ,Male ,Microcephaly ,Molecular Sequence Data ,Muscle Hypotonia ,Myopia ,Obesity ,Pakistan ,Pedigree ,Phenotype ,Retinal Degeneration ,Sequence Analysis ,DNA ,Sequence Deletion ,Vesicular Transport Proteins ,Clinical Sciences ,Genetics & Heredity ,Clinical sciences - Abstract
BackgroundCohen Syndrome (COH1) is a rare autosomal recessive disorder, principally identified by ocular, neural and muscular deficits. We identified three large consanguineous Pakistani families with intellectual disability and in some cases with autistic traits.MethodsClinical assessments were performed in order to allow comparison of clinical features with other VPS13B mutations. Homozygosity mapping followed by whole exome sequencing and Sanger sequencing strategies were used to identify disease-related mutations.ResultsWe identified two novel homozygous deletion mutations in VPS13B, firstly a 1 bp deletion, NM_017890.4:c.6879delT; p.Phe2293Leufs*24, and secondly a deletion of exons 37-40, which co-segregate with affected status. In addition to COH1-related traits, autistic features were reported in a number of family members, contrasting with the "friendly" demeanour often associated with COH1. The c.6879delT mutation is present in two families from different regions of the country, but both from the Baloch sub-ethnic group, and with a shared haplotype, indicating a founder effect among the Baloch population.ConclusionWe suspect that the c.6879delT mutation may be a common cause of COH1 and similar phenotypes among the Baloch population. Additionally, most of the individuals with the c.6879delT mutation in these two families also present with autistic like traits, and suggests that this variant may lead to a distinct autistic-like COH1 subgroup.
- Published
- 2015
128. Focal adhesions are foci for tyrosine-based signal transduction via GIV/Girdin and G proteins
- Author
-
Lopez-Sanchez, Inmaculada, Kalogriopoulos, Nicholas, Lo, I-Chung, Kabir, Firooz, Midde, Krishna K, Wang, Honghui, and Ghosh, Pradipta
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Cancer ,Amino Acid Sequence ,Animals ,COS Cells ,Cell Line ,Tumor ,Cell Movement ,Focal Adhesion Kinase 1 ,Focal Adhesions ,GTP-Binding Protein alpha Subunits ,Gi-Go ,GTP-Binding Proteins ,Guanine Nucleotide Exchange Factors ,HeLa Cells ,Humans ,Microfilament Proteins ,Phosphatidylinositol 3-Kinases ,Phosphorylation ,Protein Binding ,Signal Transduction ,Tyrosine ,Vesicular Transport Proteins ,Hela Cells ,Medical and Health Sciences ,Developmental Biology ,Biochemistry and cell biology - Abstract
GIV/Girdin is a multimodular signal transducer and a bona fide metastasis-related protein. As a guanidine exchange factor (GEF), GIV modulates signals initiated by growth factors (chemical signals) by activating the G protein Gαi. Here we report that mechanical signals triggered by the extracellular matrix (ECM) also converge on GIV-GEF via β1 integrins and that focal adhesions (FAs) serve as the major hubs for mechanochemical signaling via GIV. GIV interacts with focal adhesion kinase (FAK) and ligand-activated β1 integrins. Phosphorylation of GIV by FAK enhances PI3K-Akt signaling, the integrity of FAs, increases cell-ECM adhesion, and triggers ECM-induced cell motility. Activation of Gαi by GIV-GEF further potentiates FAK-GIV-PI3K-Akt signaling at the FAs. Spatially restricted signaling via tyrosine phosphorylated GIV at the FAs is enhanced during cancer metastasis. Thus GIV-GEF serves as a unifying platform for integration and amplification of adhesion (mechanical) and growth factor (chemical) signals during cancer progression.
- Published
- 2015
129. VTP-Identifier: Vesicular Transport Proteins Identification Based on PSSM Profiles and XGBoost.
- Author
-
Gong, Yue, Dong, Benzhi, Zhang, Zixiao, Zhai, Yixiao, Gao, Bo, Zhang, Tianjiao, and Zhang, Jingyu
- Subjects
CARRIER proteins ,PROTEIN transport ,PROTEOMICS ,RECEIVER operating characteristic curves ,PATHOLOGICAL physiology ,DNA-binding proteins - Abstract
Vesicular transport proteins are related to many human diseases, and they threaten human health when they undergo pathological changes. Protein function prediction has been one of the most in-depth topics in bioinformatics. In this work, we developed a useful tool to identify vesicular transport proteins. Our strategy is to extract transition probability composition, autocovariance transformation and other information from the position-specific scoring matrix as feature vectors. EditedNearesNeighbours (ENN) is used to address the imbalance of the data set, and the Max-Relevance-Max-Distance (MRMD) algorithm is adopted to reduce the dimension of the feature vector. We used 5-fold cross-validation and independent test sets to evaluate our model. On the test set, VTP-Identifier presented a higher performance compared with GRU. The accuracy, Matthew's correlation coefficient (MCC) and area under the ROC curve (AUC) were 83.6%, 0.531 and 0.873, respectively. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
130. A novel VPS13B mutation in Cohen syndrome: a case report and review of literature
- Author
-
Sara Momtazmanesh, Elham Rayzan, Sepideh Shahkarami, Meino Rohlfs, Christoph Klein, and Nima Rezaei
- Subjects
Cohen syndrome ,Neutropenia ,Frameshift mutation ,Vesicular transport proteins ,VPS13B protein ,Internal medicine ,RC31-1245 ,Genetics ,QH426-470 - Abstract
Abstract Background Cohen syndrome, an autosomal recessive syndrome, is a rare syndrome with diverse clinical manifestations including failure to thrive, hypotonia, hypermobile joints, microcephaly, intellectual disabilities, craniofacial and limb anomalies, neutropenia and a friendly character. It is associated with mutations of the vacuolar protein sorting 13 homolog B (VPS13B) gene, which is involved in the development of the ocular, hematological and central nervous systems. This gene encodes a transmembrane protein playing a crucial role in preserving the integrity of the Golgi complex. To date, more than 150 mutations of VPS13B have been reported in over 200 Cohen syndrome patients. Missense or nonsense mutations are the most common mutations. Case presentation A 4-year-old girl, born to consanguineous parents, was referred to the pediatric clinical immunology outpatient clinic for investigation of recurrent neutropenia with a history of recurrent infections in the past year. On physical examination, she had the characteristic facial features of Cohen syndrome, developmental delay and speech disorder. She had a cheerful disposition, and her mother gave a history of feeding difficulties in her first months of life. She did not present any ophthalmologic or cardiac abnormalities. Her lab results revealed moderate neutropenia. Serum IgG, IgM, IgA and IgE levels were normal. She fulfilled the clinical diagnostic criteria for Cohen syndrome. WES revealed a novel homozygous frameshift variant in VPS13B (LRG_351t1: c.7095del; p.Ser2366AlafsTer49). Currently, she is not experiencing any severe problem, and she undergoes irregular medical treatment once her neutrophil count decreases under the normal limit. Her verbal and motor abilities have improved as a result of speech and occupational therapies. Conclusion We reported a novel homozygous frameshift variant in VPS13B (LRG_351t1: c.7095del; p.Ser2366AlafsTer49) in a 4-year-old girl with Cohen syndrome. Cohen syndrome should be considered in differential diagnosis of any child with intellectual disability and neutropenia.
- Published
- 2020
- Full Text
- View/download PDF
131. VTP-Identifier: Vesicular Transport Proteins Identification Based on PSSM Profiles and XGBoost
- Author
-
Yue Gong, Benzhi Dong, Zixiao Zhang, Yixiao Zhai, Bo Gao, Tianjiao Zhang, and Jingyu Zhang
- Subjects
protein function prediction ,vesicular transport proteins ,machine learning ,XGBoost ,position-specific scoring matrix ,Genetics ,QH426-470 - Abstract
Vesicular transport proteins are related to many human diseases, and they threaten human health when they undergo pathological changes. Protein function prediction has been one of the most in-depth topics in bioinformatics. In this work, we developed a useful tool to identify vesicular transport proteins. Our strategy is to extract transition probability composition, autocovariance transformation and other information from the position-specific scoring matrix as feature vectors. EditedNearesNeighbours (ENN) is used to address the imbalance of the data set, and the Max-Relevance-Max-Distance (MRMD) algorithm is adopted to reduce the dimension of the feature vector. We used 5-fold cross-validation and independent test sets to evaluate our model. On the test set, VTP-Identifier presented a higher performance compared with GRU. The accuracy, Matthew’s correlation coefficient (MCC) and area under the ROC curve (AUC) were 83.6%, 0.531 and 0.873, respectively.
- Published
- 2022
- Full Text
- View/download PDF
132. Palmitoylation of ULK1 by ZDHHC13 plays a crucial role in autophagy.
- Author
-
Tabata K, Imai K, Fukuda K, Yamamoto K, Kunugi H, Fujita T, Kaminishi T, Tischer C, Neumann B, Reither S, Verissimo F, Pepperkok R, Yoshimori T, and Hamasaki M
- Subjects
- Humans, Phosphorylation, HEK293 Cells, Phosphatidylinositol Phosphates metabolism, Animals, Adaptor Proteins, Signal Transducing metabolism, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Vesicular Transport metabolism, Adaptor Proteins, Vesicular Transport genetics, Protein Transport, Vesicular Transport Proteins, Autophagy-Related Protein-1 Homolog metabolism, Autophagy-Related Protein-1 Homolog genetics, Autophagy physiology, Lipoylation, Autophagy-Related Proteins metabolism, Autophagy-Related Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Intracellular Signaling Peptides and Proteins genetics, Acyltransferases metabolism, Acyltransferases genetics, Autophagosomes metabolism
- Abstract
Autophagy is a highly conserved process from yeast to mammals in which intracellular materials are engulfed by a double-membrane organelle called autophagosome and degrading materials by fusing with the lysosome. The process of autophagy is regulated by sequential recruitment and function of autophagy-related (Atg) proteins. Genetic hierarchical analyses show that the ULK1 complex comprised of ULK1-FIP200-ATG13-ATG101 translocating from the cytosol to autophagosome formation sites as a most upstream ATG factor; this translocation is critical in autophagy initiation. However, how this translocation occurs remains unclear. Here, we show that ULK1 is palmitoylated by palmitoyltransferase ZDHHC13 and translocated to the autophagosome formation site upon autophagy induction. We find that the ULK1 palmitoylation is required for autophagy initiation. Moreover, the ULK1 palmitoylated enhances the phosphorylation of ATG14L, which is required for activating PI3-Kinase and producing phosphatidylinositol 3-phosphate, one of the autophagosome membrane's lipids. Our results reveal how the most upstream ULK1 complex translocates to the autophagosome formation sites during autophagy., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
133. Diagnostic accuracy of serum biomarkers to identify giant cell arteritis in patients with polymyalgia rheumatica.
- Author
-
Ramon A, Greigert H, Goueslard K, Cladière C, Ciudad M, Ornetti P, Audia S, Maillefert JF, Bonnotte B, and Samson M
- Subjects
- Humans, Female, Male, Aged, Chemokine CXCL9 blood, Middle Aged, Aged, 80 and over, ROC Curve, Matrix Metalloproteinase 3 blood, Vesicular Transport Proteins, Giant Cell Arteritis diagnosis, Giant Cell Arteritis blood, Polymyalgia Rheumatica blood, Polymyalgia Rheumatica diagnosis, Biomarkers blood, Interleukin-6 blood
- Abstract
Introduction: Polymyalgia rheumatica (PMR) and giant cell arteritis (GCA) are frequently overlapping conditions. Unlike in GCA, vascular inflammation is absent in PMR. Therefore, serum biomarkers reflecting vascular remodelling could be used to identify GCA in cases of apparently isolated PMR., Materials and Methods: 45 patients with isolated PMR and 29 patients with PMR/GCA overlap were included. Blood samples were collected before starting glucocorticoids for all patients. Serum biomarkers reflecting systemic inflammation (interleukin-6 (IL-6), CXCL9), vascular remodelling (MMP-2, MMP-3, MMP-9) and endothelial function (sCD141, sCD146, ICAM-1, VCAM-1, vWFA2) were measured by Luminex assays., Results: Patients with GCA had higher serum levels of sCD141 (p=0.002) and CXCL9 (p=0.002) than isolated PMR. By contrast, serum levels of MMP-3 (p=0.01) and IL-6 (p=0.004) were lower in GCA than isolated PMR. The area under the curve (AUC) was calculated for sCD141, CXCL9, IL-6 and MMP-3. Separately, none of them were >0.7, but combinations revealed higher diagnostic accuracy. The CXCL9/IL-6 ratio was significantly increased in patients with GCA (p=0.0001; cut-off >32.8, AUC 0.76), while the MMP-3/sCD141 ratio was significantly lower in patients with GCA (p<0.0001; cut-off <5.3, AUC 0.79). In patients with subclinical GCA, which is the most difficult to diagnose, sCD141 and MMP-3/sCD141 ratio demonstrated high diagnostic accuracy with AUC of 0.81 and 0.77, respectively., Conclusion: Combined serum biomarkers such as CXCL9/IL-6 and MMP-3/sCD141 could help identify GCA in patients with isolated PMR. It could allow to select patients with PMR in whom complementary examinations are needed., Competing Interests: Competing interests: MS: AbbVie consulting, Argenx consulting, Boehringer Ingelheim consulting, GSK consulting, Novartis consulting and research grant, Roche–Chugai consulting, CSL Vifor consulting, Fresenius consulting. BB: Roche–Chugai personal fees for consulting, Boehringer Ingelheim consulting. AR: Novartis research grant, Novartis consulting, AbbVie consulting, Boehringer Ingelheim consulting, Chugai consulting., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
134. ATG10-dependent autophagy is required for DDX10 to regulate cell proliferation, apoptosis and stemness in colorectal cancer.
- Author
-
Wang K, Zhan H, Fan S, Chu S, Xu H, and Jiang H
- Subjects
- Humans, Cell Line, Tumor, Ubiquitin-Conjugating Enzymes metabolism, Ubiquitin-Conjugating Enzymes genetics, Mice, Animals, Vesicular Transport Proteins, Colorectal Neoplasms pathology, Colorectal Neoplasms metabolism, Colorectal Neoplasms genetics, Autophagy physiology, Apoptosis, Cell Proliferation, DEAD-box RNA Helicases genetics, DEAD-box RNA Helicases metabolism, Autophagy-Related Proteins metabolism, Autophagy-Related Proteins genetics, Neoplastic Stem Cells pathology, Neoplastic Stem Cells metabolism
- Abstract
Colorectal cancer (CRC) remains a highly prevalent gastrointestinal neoplasm, presenting significant prevalence and lethality rate. DEAD/H box RNA helicase 10 (DDX10) has been proposed as a potential oncogene in CRC, the specific action mechanism by which DDX10 modulates the aggressive biological cellular events in CRC remains implicitly elucidated, however. During this study, DDX10 expression was detected via RT-qPCR and Western blotting. Cell proliferation was estimated via EDU staining. TUNEL staining and Western blotting appraised cell apoptosis. Cell stemness was evaluated by sphere formation assay, RT-qPCR, Western blotting as well as immunofluorescence staining. Relevant assay kit examined aldehyde dehydrogenase (ALDH) activity. Western blotting and immunofluorescence staining also detected autophagy. DDX10 was hyper-expressed in CRC cells. Down-regulation of DDX10 hampered cell proliferation, aggravated the apoptosis while eliminated the ability to form spheroid cells in CRC. In addition, DDX10 deletion improved ATG10 expression and therefore activated autophagy in CRC cells. Consequently, ATG10 depletion or treatment with autophagy inhibitor 3-Methyladenine (3-MA) partially compensated the influences of DDX10 silencing on the proliferation, apoptosis and stemness of CRC cells. Accordingly, DDX10 deficiency may aggravate autophagy mediated by ATG10 to impede cell proliferation, stemness and facilitate cell apoptosis, hence blocking the progression of CRC., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
135. The Role of ATG9 Vesicles in Autophagosome Biogenesis.
- Author
-
Holzer E, Martens S, and Tulli S
- Subjects
- Humans, Animals, Golgi Apparatus metabolism, Saccharomyces cerevisiae Proteins, Vesicular Transport Proteins, Autophagosomes metabolism, Autophagy-Related Proteins metabolism, Autophagy-Related Proteins genetics, Autophagy, Membrane Proteins metabolism
- Abstract
Autophagy mediates the degradation and recycling of cellular material in the lysosomal system. Dysfunctional autophagy is associated with a plethora of diseases including uncontrolled infections, cancer and neurodegeneration. In macroautophagy (hereafter autophagy) this material is encapsulated in double membrane vesicles, the autophagosomes, which form upon induction of autophagy. The precursors to autophagosomes, referred to as phagophores, first appear as small flattened membrane cisternae, which gradually enclose the cargo material as they grow. The assembly of phagophores during autophagy initiation has been a major subject of investigation over the past decades. A special focus has been ATG9, the only conserved transmembrane protein among the core machinery. The majority of ATG9 localizes to small Golgi-derived vesicles. Here we review the recent advances and breakthroughs regarding our understanding of how ATG9 and the vesicles it resides in serve to assemble the autophagy machinery and to establish membrane contact sites for autophagosome biogenesis. We also highlight open questions in the field that need to be addressed in the years to come., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
136. Characterization of a novel TFG variant causing autosomal recessive pure hereditary spastic paraplegia.
- Author
-
Hsiao CT, Tsai TY, Shen TY, Tsai YS, Liao YC, Lee YC, and Tsai PC
- Subjects
- Humans, Male, Female, Pedigree, Adult, Adolescent, Mutation, Young Adult, Vesicular Transport Proteins, Spastic Paraplegia, Hereditary genetics
- Abstract
Objective: TFG mutations have previously been implicated in autosomal recessive hereditary spastic paraplegia (HSP), also known as SPG57. This study aimed to investigate the clinical and molecular features of TFG mutations in a Taiwanese HSP cohort., Methods: Genetic analysis of TFG was conducted in 242 unrelated Taiwanese HSP patients using a targeted resequencing panel covering the entire coding regions of TFG. Functional assays were performed using an in vitro cell model to assess the impact of TFG variants on protein function. Additionally, other representative TFG mutant proteins were examined to understand the broader implications of TFG mutations in HSP., Results: The study identified a novel homozygous TFG c.177A>C (p.(Lys59Asn)) variant in a family with adolescent-onset, pure form HSP. Functional analysis revealed that the Lys59Asn TFG variant, similar to other HSP-associated TFG mutants, exhibited a low affinity between TFG monomers and abnormal assembly of TFG homo-oligomers. These structural alterations led to aberrant intracellular distribution, compromising TFG's protein secretion function and resulting in decreased cellular viability., Interpretation: These findings confirm that the homozygous TFG c.177A>C (p.(Lys59Asn)) variant is a novel cause of SPG57. The study expands our understanding of the clinical and mutational spectrum of TFG-associated diseases, highlighting the functional defects associated with this specific TFG variant. Overall, this research contributes to the broader comprehension of the genetic and molecular mechanisms underlying HSP., (© 2024 The Author(s). Annals of Clinical and Translational Neurology published by Wiley Periodicals LLC on behalf of American Neurological Association.)
- Published
- 2024
- Full Text
- View/download PDF
137. Heterozygous large deletion mimicking homozygous substitution in MCFD2 in a patient with combined Factor V and Factor VIII deficiency.
- Author
-
Rezigue H, Chamouni P, Fretigny M, Barbay V, Le Cam-Duchez V, Bobee V, Lanne S, Dumesnil C, Vinciguerra C, Schneider P, and Jourdy Y
- Subjects
- Humans, Male, Sequence Deletion, Female, Vesicular Transport Proteins, Factor V Deficiency genetics, Hemophilia A genetics, Heterozygote, Homozygote
- Published
- 2024
- Full Text
- View/download PDF
138. FREE1 regulates phagophore closure in plants.
- Author
-
Zhu Y, Zeng Y, and Jiang L
- Subjects
- Autophagosomes metabolism, Autophagy physiology, Endosomal Sorting Complexes Required for Transport metabolism, Phosphorylation, Protein Serine-Threonine Kinases metabolism, Signal Transduction, Vesicular Transport Proteins, Arabidopsis metabolism, Arabidopsis genetics, Arabidopsis Proteins metabolism, Arabidopsis Proteins genetics
- Abstract
Adenosine monophosphate-activated protein kinase (AMPK), the central energy sensor in more complex eukaryotes, can activate macroautophagy/autophagy upon cellular energy deficiency. However, the regulatory role of nutrient sensing in mediating phagophore closure to generate an autophagosome remains unknown. The evolutionarily conserved endosomal sorting complexes required for transport (ESCRT) machinery has been postulated to regulate phagophore sealing, yet the signaling pathway modulating the ESCRT complex relocation from multivesicular body (MVB) to phagophore for closure remains unknown. We recently identified a plant unique pleiotropic protein FREE1 (FYVE DOMAIN PROTEIN REQUIRED FOR ENDOSOMAL SORTING 1), which is phosphorylated by the plant energy sensor SnRK1 (SNF1-related kinase 1) and bridges the ATG conjugation system and ESCRT machinery to regulate phagophore sealing upon nutrient starvation. This study elucidated the bona fide roles and underlying mechanism of cellular energy-sensing pathways in regulating compartment sealing.
- Published
- 2024
- Full Text
- View/download PDF
139. Unique COPII component AtSar1a/AtSec23a pair is required for the distinct function of protein ER export in Arabidopsis thaliana
- Author
-
Zeng, Yonglun, Chung, Kin Pan, Li, Baiying, Lai, Ching Man, Lam, Sheung Kwan, Wang, Xiangfeng, Cui, Yong, Gao, Caiji, Luo, Ming, Wong, Kam-Bo, Schekman, Randy, and Jiang, Liwen
- Subjects
Genetics ,Underpinning research ,1.1 Normal biological development and functioning ,Generic health relevance ,Arabidopsis ,Arabidopsis Proteins ,Endoplasmic Reticulum ,Endoplasmic Reticulum Stress ,Models ,Molecular ,Mutation ,Missense ,Plants ,Genetically Modified ,Protein Transport ,Vesicular Transport Proteins ,coat protein complex II ,Sar1 ,Sec23 ,ER export ,functional diversity - Abstract
Secretory proteins traffic from endoplasmic reticulum (ER) to Golgi via the coat protein complex II (COPII) vesicle, which consists of five cytosolic components (Sar1, Sec23-24, and Sec13-31). In eukaryotes, COPII transport has diversified due to gene duplication, creating multiple COPII paralogs. Evidence has accumulated, revealing the functional heterogeneity of COPII paralogs in protein ER export. Sar1B, the small GTPase of COPII machinery, seems to be specialized for large cargo secretion in mammals. Arabidopsis contains five Sar1 and seven Sec23 homologs, and AtSar1a was previously shown to exhibit different effects on α-amylase secretion. However, mechanisms underlying the functional diversity of Sar1 paralogs remain unclear in higher organisms. Here, we show that the Arabidopsis Sar1 homolog AtSar1a exhibits distinct localization in plant cells. Transgenic Arabidopsis plants expressing dominant-negative AtSar1a exhibit distinct effects on ER cargo export. Mutagenesis analysis identified a single amino acid, Cys84, as being responsible for the functional diversity of AtSar1a. Structure homology modeling and interaction studies revealed that Cys84 is crucial for the specific interaction of AtSar1a with AtSec23a, a distinct Arabidopsis Sec23 homolog. Structure modeling and coimmunoprecipitation further identified a corresponding amino acid, Cys484, on AtSec23a as being essential for the specific pair formation. At the cellular level, the Cys484 mutation affects the distinct function of AtSec23a on vacuolar cargo trafficking. Additionally, dominant-negative AtSar1a affects the ER export of the transcription factor bZIP28 under ER stress. We have demonstrated a unique plant pair of COPII machinery function in ER export and the mechanism underlying the functional diversity of COPII paralogs in eukaryotes.
- Published
- 2015
140. Activation of G proteins by GIV-GEF is a pivot point for insulin resistance and sensitivity
- Author
-
S., Gary, Lopez-Sanchez, Inmaculada, Aznar, Nicolas, Kalogriopoulos, Nicholas, Pedram, Shabnam, Midde, Krishna, Ciaraldi, Theodore P, Henry, Robert R, and Ghosh, Pradipta
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Diabetes ,Genetics ,Metabolic and endocrine ,Cells ,Cultured ,Fatty Acids ,Female ,GTP-Binding Protein Regulators ,GTP-Binding Protein alpha Subunits ,Gi-Go ,Guanine Nucleotide Exchange Factors ,Humans ,Insulin ,Insulin Receptor Substrate Proteins ,Insulin Resistance ,Microfilament Proteins ,Muscle ,Skeletal ,Phosphatidylinositol 3-Kinase ,Phosphorylation ,Protein-Tyrosine Kinases ,Receptor ,Insulin ,Signal Transduction ,Vesicular Transport Proteins ,Medical and Health Sciences ,Developmental Biology ,Biochemistry and cell biology - Abstract
Insulin resistance (IR) is a metabolic disorder characterized by impaired insulin signaling and cellular glucose uptake. The current paradigm for insulin signaling centers upon the insulin receptor (InsR) and its substrate IRS1; the latter is believed to be the sole conduit for postreceptor signaling. Here we challenge that paradigm and show that GIV/Girdin, a guanidine exchange factor (GEF) for the trimeric G protein Gαi, is another major hierarchical conduit for the metabolic insulin response. By virtue of its ability to directly bind InsR, IRS1, and phosphoinositide 3-kinase, GIV serves as a key hub in the immediate postreceptor level, which coordinately enhances the metabolic insulin response and glucose uptake in myotubes via its GEF function. Site-directed mutagenesis or phosphoinhibition of GIV-GEF by the fatty acid/protein kinase C-theta pathway triggers IR. Insulin sensitizers reverse phosphoinhibition of GIV and reinstate insulin sensitivity. We also provide evidence for such reversible regulation of GIV-GEF in skeletal muscles from patients with IR. Thus GIV is an essential upstream component that couples InsR to G-protein signaling to enhance the metabolic insulin response, and impairment of such coupling triggers IR. We also provide evidence that GIV-GEF serves as therapeutic target for exogenous manipulation of physiological insulin response and reversal of IR in skeletal muscles.
- Published
- 2015
141. Activation of G proteins by GIV-GEF is a pivot point for insulin resistance and sensitivity.
- Author
-
Ma, Gary S, Lopez-Sanchez, Inmaculada, Aznar, Nicolas, Kalogriopoulos, Nicholas, Pedram, Shabnam, Midde, Krishna, Ciaraldi, Theodore P, Henry, Robert R, and Ghosh, Pradipta
- Subjects
Muscle ,Skeletal ,Cells ,Cultured ,Humans ,Insulin Resistance ,Microfilament Proteins ,Insulin ,GTP-Binding Protein alpha Subunits ,Gi-Go ,Receptor ,Insulin ,Fatty Acids ,GTP-Binding Protein Regulators ,Guanine Nucleotide Exchange Factors ,Vesicular Transport Proteins ,Signal Transduction ,Phosphorylation ,Female ,Protein-Tyrosine Kinases ,Insulin Receptor Substrate Proteins ,Phosphatidylinositol 3-Kinase ,Muscle ,Skeletal ,Cells ,Cultured ,GTP-Binding Protein alpha Subunits ,Gi-Go ,Receptor ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
Insulin resistance (IR) is a metabolic disorder characterized by impaired insulin signaling and cellular glucose uptake. The current paradigm for insulin signaling centers upon the insulin receptor (InsR) and its substrate IRS1; the latter is believed to be the sole conduit for postreceptor signaling. Here we challenge that paradigm and show that GIV/Girdin, a guanidine exchange factor (GEF) for the trimeric G protein Gαi, is another major hierarchical conduit for the metabolic insulin response. By virtue of its ability to directly bind InsR, IRS1, and phosphoinositide 3-kinase, GIV serves as a key hub in the immediate postreceptor level, which coordinately enhances the metabolic insulin response and glucose uptake in myotubes via its GEF function. Site-directed mutagenesis or phosphoinhibition of GIV-GEF by the fatty acid/protein kinase C-theta pathway triggers IR. Insulin sensitizers reverse phosphoinhibition of GIV and reinstate insulin sensitivity. We also provide evidence for such reversible regulation of GIV-GEF in skeletal muscles from patients with IR. Thus GIV is an essential upstream component that couples InsR to G-protein signaling to enhance the metabolic insulin response, and impairment of such coupling triggers IR. We also provide evidence that GIV-GEF serves as therapeutic target for exogenous manipulation of physiological insulin response and reversal of IR in skeletal muscles.
- Published
- 2015
142. Cyclin-dependent kinase 5 activates guanine nucleotide exchange factor GIV/Girdin to orchestrate migration–proliferation dichotomy
- Author
-
Bhandari, Deepali, Lopez-Sanchez, Inmaculada, To, Andrew, Lo, I-Chung, Aznar, Nicolas, Leyme, Anthony, Gupta, Vijay, Niesman, Ingrid, Maddox, Adam L, Garcia-Marcos, Mikel, Farquhar, Marilyn G, and Ghosh, Pradipta
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Cancer ,Underpinning research ,1.1 Normal biological development and functioning ,Amino Acid Sequence ,Animals ,Cell Movement ,Cell Proliferation ,Cyclin-Dependent Kinase 5 ,ErbB Receptors ,Humans ,Microfilament Proteins ,Molecular Sequence Data ,Morphogenesis ,Phosphorylation ,Protein Transport ,Sequence Homology ,Amino Acid ,Signal Transduction ,Vesicular Transport Proteins ,Wound Healing ,migration-proliferation dichotomy ,heterotrimeric G protein ,growth factor receptor tyrosine kinase ,GIV/Girdin ,guanine nucleotide exchange factor ,migration–proliferation dichotomy - Abstract
Signals propagated by receptor tyrosine kinases (RTKs) can drive cell migration and proliferation, two cellular processes that do not occur simultaneously--a phenomenon called "migration-proliferation dichotomy." We previously showed that epidermal growth factor (EGF) signaling is skewed to favor migration over proliferation via noncanonical transactivation of Gαi proteins by the guanine exchange factor (GEF) GIV. However, what turns on GIV-GEF downstream of growth factor RTKs remained unknown. Here we reveal the molecular mechanism by which phosphorylation of GIV by cyclin-dependent kinase 5 (CDK5) triggers GIV's ability to bind and activate Gαi in response to growth factors and modulate downstream signals to establish a dichotomy between migration and proliferation. We show that CDK5 binds and phosphorylates GIV at Ser1674 near its GEF motif. When Ser1674 is phosphorylated, GIV activates Gαi and enhances promigratory Akt signals. Phosphorylated GIV also binds Gαs and enhances endosomal maturation, which shortens the transit time of EGFR through early endosomes, thereby limiting mitogenic MAPK signals. Consequently, this phosphoevent triggers cells to preferentially migrate during wound healing and transmigration of cancer cells. When Ser1674 cannot be phosphorylated, GIV cannot bind either Gαi or Gαs, Akt signaling is suppressed, mitogenic signals are enhanced due to delayed transit time of EGFR through early endosomes, and cells preferentially proliferate. These results illuminate how GIV-GEF is turned on upon receptor activation, adds GIV to the repertoire of CDK5 substrates, and defines a mechanism by which this unusual CDK orchestrates migration-proliferation dichotomy during cancer invasion, wound healing, and development.
- Published
- 2015
143. Therapeutic effects of cell-permeant peptides that activate G proteins downstream of growth factors
- Author
-
S., Gary, Aznar, Nicolas, Kalogriopoulos, Nicholas, Midde, Krishna K, Lopez-Sanchez, Inmaculada, Sato, Emi, Dunkel, Ying, Gallo, Richard L, and Ghosh, Pradipta
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Genetics ,Underpinning research ,1.1 Normal biological development and functioning ,Animals ,Cell-Penetrating Peptides ,Fluorescence Resonance Energy Transfer ,GTP-Binding Proteins ,Gene Products ,tat ,Genetic Engineering ,HeLa Cells ,Humans ,Intercellular Signaling Peptides and Proteins ,Mice ,Microfilament Proteins ,Models ,Molecular ,Polymerase Chain Reaction ,Signal Transduction ,Transduction ,Genetic ,Vesicular Transport Proteins ,heterotrimeric G proteins ,fibrosis ,cell-permeable GIV/Girdin peptide ,wound healing ,invasion ,Hela Cells - Abstract
In eukaryotes, receptor tyrosine kinases (RTKs) and trimeric G proteins are two major signaling hubs. Signal transduction via trimeric G proteins has long been believed to be triggered exclusively by G protein-coupled receptors (GPCRs). This paradigm has recently been challenged by several studies on a multimodular signal transducer, Gα-Interacting Vesicle associated protein (GIV/Girdin). We recently demonstrated that GIV's C terminus (CT) serves as a platform for dynamic association of ligand-activated RTKs with Gαi, and for noncanonical transactivation of G proteins. However, exogenous manipulation of this platform has remained beyond reach. Here we developed cell-permeable GIV-CT peptides by fusing a TAT-peptide transduction domain (TAT-PTD) to the minimal modular elements of GIV that are necessary and sufficient for activation of Gi downstream of RTKs, and used them to engineer signaling networks and alter cell behavior. In the presence of an intact GEF motif, TAT-GIV-CT peptides enhanced diverse processes in which GIV's GEF function has previously been implicated, e.g., 2D cell migration after scratch-wounding, invasion of cancer cells, and finally, myofibroblast activation and collagen production. Furthermore, topical application of TAT-GIV-CT peptides enhanced the complex, multireceptor-driven process of wound repair in mice in a GEF-dependent manner. Thus, TAT-GIV peptides provide a novel and versatile tool to manipulate Gαi activation downstream of growth factors in a diverse array of pathophysiologic conditions.
- Published
- 2015
144. Activation of Gαi at the Golgi by GIV/Girdin Imposes Finiteness in Arf1 Signaling
- Author
-
Lo, I-Chung, Gupta, Vijay, Midde, Krishna K, Taupin, Vanessa, Lopez-Sanchez, Inmaculada, Kufareva, Irina, Abagyan, Ruben, Randazzo, Paul A, Farquhar, Marilyn G, and Ghosh, Pradipta
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,1.1 Normal biological development and functioning ,Underpinning research ,Generic health relevance ,ADP-Ribosylation Factor 1 ,ADP-Ribosylation Factors ,Animals ,Binding Sites ,COS Cells ,Cell Line ,Cell Membrane ,Chlorocebus aethiops ,Coatomer Protein ,Enzyme Activation ,GTP-Binding Protein alpha Subunits ,Gi-Go ,GTPase-Activating Proteins ,Golgi Apparatus ,HEK293 Cells ,HeLa Cells ,Humans ,Microfilament Proteins ,Protein Binding ,Protein Structure ,Tertiary ,Protein Transport ,RNA Interference ,RNA ,Small Interfering ,Signal Transduction ,Transport Vesicles ,Vesicular Transport Proteins ,Hela Cells ,Medical and Health Sciences ,Developmental Biology ,Biochemistry and cell biology - Abstract
A long-held tenet of heterotrimeric G protein signal transduction is that it is triggered by G protein-coupled receptors (GPCRs) at the PM. Here, we demonstrate that Gi is activated in the Golgi by GIV/Girdin, a non-receptor guanine-nucleotide exchange factor (GEF). GIV-dependent activation of Gi at the Golgi maintains the finiteness of the cyclical activation of ADP-ribosylation factor 1 (Arf1), a fundamental step in vesicle traffic in all eukaryotes. Several interactions with other major components of Golgi trafficking-e.g., active Arf1, its regulator, ArfGAP2/3, and the adaptor protein β-COP-enable GIV to coordinately regulate Arf1 signaling. When the GIV-Gαi pathway is selectively inhibited, levels of GTP-bound Arf1 are elevated and protein transport along the secretory pathway is delayed. These findings define a paradigm in non-canonical G protein signaling at the Golgi, which places GIV-GEF at the crossroads between signals gated by the trimeric G proteins and the Arf family of monomeric GTPases.
- Published
- 2015
145. Multimodular biosensors reveal a novel platform for activation of G proteins by growth factor receptors
- Author
-
Midde, Krishna K, Aznar, Nicolas, Laederich, Melanie B, S., Gary, Kunkel, Maya T, Newton, Alexandra C, and Ghosh, Pradipta
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Emerging Infectious Diseases ,Bioengineering ,1.1 Normal biological development and functioning ,Underpinning research ,Generic health relevance ,Animals ,Biosensing Techniques ,COS Cells ,Chlorocebus aethiops ,Fluorescence Resonance Energy Transfer ,GTP-Binding Proteins ,Humans ,Microfilament Proteins ,Receptor Protein-Tyrosine Kinases ,Receptors ,Growth Factor ,Signal Transduction ,Vesicular Transport Proteins ,heterotrimeric G protein ,growth factor receptor tyrosine kinase ,Girdin ,PI3-kinase ,Akt ,invasion ,cyclic AMP - Abstract
Environmental cues are transmitted to the interior of the cell via a complex network of signaling hubs. Receptor tyrosine kinases (RTKs) and trimeric G proteins are two such major signaling hubs in eukaryotes. Conventionally, canonical signal transduction via trimeric G proteins is thought to be triggered exclusively by G protein-coupled receptors. Here we used molecular engineering to develop modular fluorescent biosensors that exploit the remarkable specificity of bimolecular recognition, i.e., of both G proteins and RTKs, and reveal the workings of a novel platform for activation of G proteins by RTKs in single living cells. Comprised of the unique modular makeup of guanidine exchange factor Gα-interacting vesicle-associated protein (GIV)/girdin, a guanidine exchange factor that links G proteins to a variety of RTKs, these biosensors provide direct evidence that RTK-GIV-Gαi ternary complexes are formed in living cells and that Gαi is transactivated within minutes after growth factor stimulation at the plasma membrane. Thus, GIV-derived biosensors provide a versatile strategy for visualizing, monitoring, and manipulating the dynamic association of Gαi with RTKs for noncanonical transactivation of G proteins in cells and illuminate a fundamental signaling event regulated by GIV during diverse cellular processes and pathophysiologic states.
- Published
- 2015
146. Ciliary Vesicle Formation: A Prelude to Ciliogenesis
- Author
-
Yee, Laura E and Reiter, Jeremy F
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Pediatric ,Animals ,Carrier Proteins ,Cilia ,Epithelial Cells ,Gene Expression Regulation ,Developmental ,Humans ,Transport Vesicles ,Vesicular Transport Proteins ,Medical and Health Sciences ,Developmental Biology ,Biochemistry and cell biology - Abstract
Reporting recently in Nature Cell Biology, Lu et al. (2015) identify two Eps15-homology-domain-containing proteins as critical effectors of ciliary vesicle formation, an early event in ciliogenesis. Functional dissection reveals that one of them works to convert small vesicles associated with mother centriole distal appendages into a larger ciliary vesicle.
- Published
- 2015
147. GIV/Girdin Transmits Signals from Multiple Receptors by Triggering Trimeric G Protein Activation*
- Author
-
Garcia-Marcos, Mikel, Ghosh, Pradipta, and Farquhar, Marilyn G
- Subjects
Pharmacology and Pharmaceutical Sciences ,Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Animals ,Cell Movement ,Cell Survival ,GTP-Binding Proteins ,Humans ,Liver Cirrhosis ,Microfilament Proteins ,Mitosis ,Models ,Molecular ,Neoplasm Metastasis ,Neoplasms ,Nephrotic Syndrome ,Protein Multimerization ,Receptors ,Cell Surface ,Signal Transduction ,Vesicular Transport Proteins ,Guanine Nucleotide Exchange Factor ,Heterotrimeric G Protein ,Liver Fibrosis ,Receptor Tyrosine Kinase ,Src Homology 2 Domain ,Tumor Metastasis ,Chemical Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
Activation of trimeric G proteins has been traditionally viewed as the exclusive job of G protein-coupled receptors (GPCRs). This view has been challenged by the discovery of non-receptor activators of trimeric G proteins. Among them, GIV (a.k.a. Girdin) is the first for which a guanine nucleotide exchange factor (GEF) activity has been unequivocally associated with a well defined motif. Here we discuss how GIV assembles alternative signaling pathways by sensing cues from various classes of surface receptors and relaying them via G protein activation. We also describe the dysregulation of this mechanism in disease and how its targeting holds promise for novel therapeutics.
- Published
- 2015
148. A Combined Proteomics/Genomics Approach Links Hepatitis C Virus Infection with Nonsense-Mediated mRNA Decay
- Author
-
Ramage, Holly R, Kumar, G Renuka, Verschueren, Erik, Johnson, Jeffrey R, Von Dollen, John, Johnson, Tasha, Newton, Billy, Shah, Priya, Horner, Julie, Krogan, Nevan J, and Ott, Melanie
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Biological Sciences ,Genetics ,Emerging Infectious Diseases ,Digestive Diseases ,Biotechnology ,Chronic Liver Disease and Cirrhosis ,Hepatitis - C ,Liver Disease ,Hepatitis ,Infectious Diseases ,2.2 Factors relating to the physical environment ,2.1 Biological and endogenous factors ,Aetiology ,Infection ,Good Health and Well Being ,Carrier Proteins ,Cell Line ,Tumor ,Hepacivirus ,Hepatitis C ,Host-Pathogen Interactions ,Humans ,Intracellular Signaling Peptides and Proteins ,Mitochondria ,Mitochondrial Proteins ,Nonsense Mediated mRNA Decay ,Protein Interaction Maps ,Protein Transport ,Proteome ,Proteomics ,Vesicular Transport Proteins ,Viral Core Proteins ,Viral Nonstructural Proteins ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
Hepatitis C virus (HCV) is a leading cause of liver disease, but insight into virus-host interactions remains limited. We systematically used affinity purification/mass spectrometry to define the host interactions of all ten HCV proteins in hepatoma cells. We combined these studies with RNAi knockdown of corresponding genes using a two-step scoring approach to generate a map of 139 high-confidence HCV-host protein-protein interactions. We found mitochondrial proteins highly involved in HCV infection and characterized an interaction between the viral core protein and host protein within bgcn homolog (WIBG). Expression of core prevents WIBG from binding its regular interaction partners Y14 and Magoh, two known mediators of the nonsense-mediated mRNA decay pathway. We discovered that this surveillance pathway is disrupted in HCV-infected cells, causing potentially harmful transcripts to accumulate. Our study provides a comprehensive view of HCV-host interactions and uncovers mechanisms for how HCV perturbs host functions during infection.
- Published
- 2015
149. Multiple functions of the SNARE protein Snap29 in autophagy, endocytic, and exocytic trafficking during epithelial formation in Drosophila
- Author
-
Morelli, Elena, Ginefra, Pierpaolo, Mastrodonato, Valeria, Beznoussenko, Galina V, Rusten, Tor Erik, Bilder, David, Stenmark, Harald, Mironov, Alexandre A, and Vaccari, Thomas
- Subjects
Genetics ,Underpinning research ,1.1 Normal biological development and functioning ,Animals ,Autophagy ,Cell Movement ,Drosophila Proteins ,Drosophila melanogaster ,Endosomes ,Exosomes ,Humans ,Phagosomes ,Protein Binding ,Protein Transport ,Qb-SNARE Proteins ,Qc-SNARE Proteins ,SNARE Proteins ,Vesicular Transport Proteins ,autophagy ,dome ,Notch ,Snap29 ,SNARE ,trafficking ,usnp ,Atg ,autophagy-related ,CEDNIK ,cerebral dysgenesis ,neuropathy ,ichthyosis ,and palmoplantar keratoderma ,CFP ,cyan fluorescent protein ,E(spl)mβ-HLH ,enhancer of split mβ ,helix-loop-helix ,EM ,electron microscopy ,ESCRT ,endosomal sorting complex required for transport ,FE ,follicular epithelium ,GFP ,green fluorescent protein ,MENE ,mutant eye no eclosion ,MVB ,multivesicular body ,N ,Notch ,NECD ,N extracellular domain ,NPF ,asparagine-proline-phenylalanine ,SNARE ,soluble NSF attachment protein receptor ,Snap29 ,synaptosomal-associated protein 29 kDa ,Socs36E ,suppressor of cytokine signaling at 36E ,Syb ,Synaptobrevin ,Syx ,syntaxin ,V-ATPase ,vacuolar H+-ATPase ,Vamp ,vesicle-associated membrane protein ,Vps25 ,vacuolar protein sorting 25 ,WT ,wild type ,dome ,domeless ,histone H3 ,His3 ,hop-Stat92E ,hopscotch-signal transducer and activator of transcription protein at 92E ,os ,outstretched ,ref(2)P ,refractory to sigma P ,Biochemistry and Cell Biology ,Biochemistry & Molecular Biology - Abstract
How autophagic degradation is linked to endosomal trafficking routes is little known. Here we screened a collection of uncharacterized Drosophila mutants affecting membrane transport to identify new genes that also have a role in autophagy. We isolated a loss of function mutant in Snap29 (Synaptosomal-associated protein 29 kDa), the gene encoding the Drosophila homolog of the human protein SNAP29 and have characterized its function in vivo. Snap29 contains 2 soluble NSF attachment protein receptor (SNARE) domains and a asparagine-proline-phenylalanine (NPF motif) at its N terminus and rescue experiments indicate that both SNARE domains are required for function, whereas the NPF motif is in part dispensable. We find that Snap29 interacts with SNARE proteins, localizes to multiple trafficking organelles, and is required for protein trafficking and for proper Golgi apparatus morphology. Developing tissue lacking Snap29 displays distinctive epithelial architecture defects and accumulates large amounts of autophagosomes, highlighting a major role of Snap29 in autophagy and secretion. Mutants for autophagy genes do not display epithelial architecture or secretion defects, suggesting that the these alterations of the Snap29 mutant are unlikely to be caused by the impairment of autophagy. In contrast, we find evidence of elevated levels of hop-Stat92E (hopscotch-signal transducer and activator of transcription protein at 92E) ligand, receptor, and associated signaling, which might underlie the epithelial defects. In summary, our findings support a role of Snap29 at key steps of membrane trafficking, and predict that signaling defects may contribute to the pathogenesis of cerebral dysgenesis, neuropathy, ichthyosis, and palmoplantar keratoderma (CEDNIK), a human congenital syndrome due to loss of Snap29.
- Published
- 2014
150. Chronic enrichment of hepatic endoplasmic reticulum-mitochondria contact leads to mitochondrial dysfunction in obesity.
- Author
-
Pers, Benedicte, Parlakgül, Güneş, Güney, Ekin, Inouye, Karen, Hotamisligil, Gökhan, and Arruda, Ana Paula
- Subjects
Animals ,Calcium ,Calnexin ,Disease Models ,Animal ,Down-Regulation ,Endoplasmic Reticulum ,Endoplasmic Reticulum Stress ,GTP Phosphohydrolases ,Glucose ,Hepatocytes ,Inositol 1 ,4 ,5-Trisphosphate Receptors ,Lipid Metabolism ,Liver ,Mice ,Microscopy ,Electron ,Transmission ,Mitochondria ,Obesity ,Oxidative Stress ,Vesicular Transport Proteins - Abstract
Proper function of the endoplasmic reticulum (ER) and mitochondria is crucial for cellular homeostasis, and dysfunction at either site has been linked to pathophysiological states, including metabolic diseases. Although the ER and mitochondria play distinct cellular roles, these organelles also form physical interactions with each other at sites defined as mitochondria-associated ER membranes (MAMs), which are essential for calcium, lipid and metabolite exchange. Here we show that in the liver, obesity leads to a marked reorganization of MAMs resulting in mitochondrial calcium overload, compromised mitochondrial oxidative capacity and augmented oxidative stress. Experimental induction of ER-mitochondria interactions results in oxidative stress and impaired metabolic homeostasis, whereas downregulation of PACS-2 or IP3R1, proteins important for ER-mitochondria tethering or calcium transport, respectively, improves mitochondrial oxidative capacity and glucose metabolism in obese animals. These findings establish excessive ER-mitochondrial coupling as an essential component of organelle dysfunction in obesity that may contribute to the development of metabolic pathologies such as insulin resistance and diabetes.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.