3,244 results on '"Protein Transport genetics"'
Search Results
2. The cullin Rtt101 promotes ubiquitin-dependent DNA-protein crosslink repair across the cell cycle.
- Author
-
Noireterre A, Soudet J, Bagdiul I, and Stutz F
- Subjects
- Saccharomyces cerevisiae, DNA Adducts metabolism, DNA Topoisomerases, Type I metabolism, Protein Transport genetics, Ubiquitination genetics, DNA Replication genetics, Multienzyme Complexes metabolism, Cullin Proteins genetics, Cullin Proteins metabolism, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins metabolism, Cell Cycle genetics, DNA Repair
- Abstract
DNA-protein crosslinks (DPCs) challenge faithful DNA replication and smooth passage of genomic information. Our study unveils the cullin E3 ubiquitin ligase Rtt101 as a DPC repair factor. Genetic analyses demonstrate that Rtt101 is essential for resistance to a wide range of DPC types including topoisomerase 1 crosslinks, in the same pathway as the ubiquitin-dependent aspartic protease Ddi1. Using an in vivo inducible Top1-mimicking DPC system, we reveal the significant impact of Rtt101 ubiquitination on DPC removal across different cell cycle phases. High-throughput methods coupled with next-generation sequencing specifically highlight the association of Rtt101 with replisomes as well as colocalization with DPCs. Our findings establish Rtt101 as a main contributor to DPC repair throughout the yeast cell cycle., (© The Author(s) 2024. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2024
- Full Text
- View/download PDF
3. Integrative analysis of Lunatic Fringe variants associated with spondylocostal dysostosis type-III.
- Author
-
Wengryn P, Fenrich F, Silveira KDC, Oborn C, Mizumoto S, Beke A, Soltys CL, Yamada S, and Kannu P
- Subjects
- Animals, Mice, Cell Line, Chlorocebus aethiops, Genomics, NIH 3T3 Cells, Protein Processing, Post-Translational genetics, Protein Transport genetics, Proteomics, Dysostoses congenital, Dysostoses genetics, Genetic Variation genetics, Glycosyltransferases genetics
- Abstract
Lunatic Fringe (LFNG) is required for spinal development. Biallelic pathogenic variants cause spondylocostal dysostosis type-III (SCD3), a rare disease generally characterized by malformed, asymmetrical, and attenuated development of the vertebral column and ribs. However, a variety of SCD3 cases reported have presented with additional features such as auditory alterations and digit abnormalities. There has yet to be a single, comprehensive, functional evaluation of causative LFNG variants and such analyses could unveil molecular mechanisms for phenotypic variability in SCD3. Therefore, nine LFNG missense variants associated with SCD3, c.564C>A, c.583T>C, c.842C>A, c.467T>G, c.856C>T, c.601G>A, c.446C>T, c.521G>A, and c.766G>A, were assessed in vitro for subcellular localization and protein processing. Glycosyltransferase activity was quantified for the first time in the c.583T>C, c.842C>A, and c.446C>T variants. Primarily, our results are the first to satisfy American College of Medical Genetics and Genomics PS3 criteria (functional evidence via well-established assay) for the pathogenicity of c.583T>C, c.842C>A, and c.446C>T, and replicate this evidence for the remaining six variants. Secondly, this work indicates that all variants that prevent Golgi localization also lead to impaired protein processing. It appears that the FRINGE domain is responsible for this phenomenon. Thirdly, our data suggests that variant proximity to the catalytic residue may influence whether LFNG is improperly trafficked and/or enzymatically dysfunctional. Finally, the phenotype of the axial skeleton, but not elsewhere, may be modulated in a variant-specific fashion. More reports are needed to continue testing this hypothesis. We anticipate our data will be used as a basis for discussion of genotype-phenotype correlations in SCD3., (© 2024 The Author(s). The FASEB Journal published by Wiley Periodicals LLC on behalf of Federation of American Societies for Experimental Biology.)
- Published
- 2024
- Full Text
- View/download PDF
4. Polarized transport requires AP-1-mediated recruitment of KIF13A and KIF13B at the trans-Golgi.
- Author
-
Montgomery AC, Mendoza CS, Garbouchian A, Quinones GB, and Bentley M
- Subjects
- Cells, Cultured, Neurons metabolism, Protein Transport genetics, Protein Transport physiology, Golgi Apparatus metabolism, Kinesins metabolism, Adaptor Protein Complex 1 metabolism, trans-Golgi Network metabolism
- Abstract
Neurons are polarized cells that require accurate membrane trafficking to maintain distinct protein complements at dendritic and axonal membranes. The Kinesin-3 family members KIF13A and KIF13B are thought to mediate dendrite-selective transport, but the mechanism by which they are recruited to polarized vesicles and the differences in the specific trafficking role of each KIF13 have not been defined. We performed live-cell imaging in cultured hippocampal neurons and found that KIF13A is a dedicated dendrite-selective kinesin. KIF13B confers two different transport modes, dendrite- and axon-selective transport. Both KIF13s are maintained at the trans-Golgi network by interactions with the heterotetrameric adaptor protein complex AP-1. Interference with KIF13 binding to AP-1 resulted in disruptions to both dendrite- and axon-selective trafficking. We propose that AP-1 is the molecular link between the sorting of polarized cargoes into vesicles and the recruitment of kinesins that confer polarized transport.
- Published
- 2024
- Full Text
- View/download PDF
5. Sphingosine-1-phosphate receptor 3 regulates the transendothelial transport of high-density lipoproteins and low-density lipoproteins in opposite ways.
- Author
-
Velagapudi S, Wang D, Poti F, Feuerborn R, Robert J, Schlumpf E, Yalcinkaya M, Panteloglou G, Potapenko A, Simoni M, Rohrer L, Nofer JR, and von Eckardstein A
- Subjects
- Animals, Humans, Mice, Biological Transport, Cells, Cultured, Disease Models, Animal, Lysophospholipids, Mice, Inbred C57BL, Mice, Knockout, ApoE, Scavenger Receptors, Class B metabolism, Scavenger Receptors, Class B genetics, Atherosclerosis metabolism, Atherosclerosis genetics, Atherosclerosis pathology, Atherosclerosis prevention & control, Endothelial Cells metabolism, Lipoproteins, HDL metabolism, Lipoproteins, LDL metabolism, Sphingosine-1-Phosphate Receptors metabolism, Sphingosine-1-Phosphate Receptors genetics, Protein Transport genetics
- Abstract
Aims: The entry of lipoproteins from blood into the arterial wall is a rate-limiting step in atherosclerosis. It is controversial whether this happens by filtration or regulated transendothelial transport.Because sphingosine-1-phosphate (S1P) preserves the endothelial barrier, we investigated in vivo and in vitro, whether S1P and its cognate S1P-receptor 3 (S1P3) regulate the transendothelial transport of lipoproteins., Methods and Results: Compared to apoE-haploinsufficient mice (CTRL), apoE-haploinsufficient mice with additional endothelium-specific knock-in of S1P3 (S1P3-iECKI) showed decreased transport of LDL and Evan's Blue but increased transport of HDL from blood into the peritoneal cave. After 30 weeks of high-fat diet feeding, S1P3-iECKI mice had lower levels of non-HDL-cholesterol and less atherosclerosis than CTRL mice. In vitro stimulation with an S1P3 agonist increased the transport of 125I-HDL but decreased the transport of 125I-LDL through human aortic endothelial cells (HAECs). Conversely, inhibition or knock-down of S1P3 decreased the transport of 125I-HDL but increased the transport of 125I-LDL. Silencing of SCARB1 encoding scavenger receptor B1 (SR-BI) abrogated the stimulation of 125I-HDL transport by the S1P3 agonist. The transendothelial transport of 125I-LDL was decreased by silencing of SCARB1 or ACVLR1 encoding activin-like kinase 1 but not by interference with LDLR. None of the three knock-downs prevented the stimulatory effect of S1P3 inhibition on transendothelial 125I-LDL transport., Conclusion: S1P3 regulates the transendothelial transport of HDL and LDL oppositely by SR-BI-dependent and SR-BI-independent mechanisms, respectively. This divergence supports a contention that lipoproteins pass the endothelial barrier by specifically regulated mechanisms rather than passive filtration., Competing Interests: Conflict of interest: none declared., (© The Author(s) 2023. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2024
- Full Text
- View/download PDF
6. Modulation of peroxisomal import by the PEX13 SH3 domain and a proximal FxxxF binding motif.
- Author
-
Gaussmann S, Peschel R, Ott J, Zak KM, Sastre J, Delhommel F, Popowicz GM, Boekhoven J, Schliebs W, Erdmann R, and Sattler M
- Subjects
- Humans, Peptides chemistry, Peroxisome-Targeting Signal 1 Receptor metabolism, Peroxisomes metabolism, Protein Binding, Saccharomyces cerevisiae metabolism, Membrane Proteins chemistry, Membrane Proteins genetics, Membrane Proteins metabolism, Protein Transport genetics, Protein Transport physiology, Saccharomyces cerevisiae Proteins metabolism, src Homology Domains genetics, src Homology Domains physiology
- Abstract
Import of proteins into peroxisomes depends on PEX5, PEX13 and PEX14. By combining biochemical methods and structural biology, we show that the C-terminal SH3 domain of PEX13 mediates intramolecular interactions with a proximal FxxxF motif. The SH3 domain also binds WxxxF peptide motifs in the import receptor PEX5, demonstrating evolutionary conservation of such interactions from yeast to human. Strikingly, intramolecular interaction of the PEX13 FxxxF motif regulates binding of PEX5 WxxxF/Y motifs to the PEX13 SH3 domain. Crystal structures reveal how FxxxF and WxxxF/Y motifs are recognized by a non-canonical surface on the SH3 domain. The PEX13 FxxxF motif also mediates binding to PEX14. Surprisingly, the potential PxxP binding surface of the SH3 domain does not recognize PEX14 PxxP motifs, distinct from its yeast ortholog. Our data show that the dynamic network of PEX13 interactions with PEX5 and PEX14, mediated by diaromatic peptide motifs, modulates peroxisomal matrix import., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
7. VPS35 and retromer dysfunction in Parkinson's disease.
- Author
-
Rowlands J and Moore DJ
- Subjects
- Animals, Humans, Vesicular Transport Proteins genetics, Vesicular Transport Proteins metabolism, Protein Transport genetics, Mutation, Parkinson Disease genetics, Parkinson Disease metabolism, Parkinson Disease pathology, Neurodegenerative Diseases
- Abstract
The vacuolar protein sorting 35 ortholog ( VPS35 ) gene encodes a core component of the retromer complex essential for the endosomal sorting and recycling of transmembrane cargo. Endo-lysosomal pathway deficits are suggested to play a role in the pathogenesis of neurodegenerative diseases, including Parkinson's disease (PD). Mutations in VPS35 cause a late-onset, autosomal dominant form of PD, with a single missense mutation (D620N) shown to segregate with disease in PD families. Understanding how the PD-linked D620N mutation causes retromer dysfunction will provide valuable insight into the pathophysiology of PD and may advance the identification of therapeutics. D620N VPS35 can induce LRRK2 hyperactivation and impair endosomal recruitment of the WASH complex but is also linked to mitochondrial and autophagy-lysosomal pathway dysfunction and altered neurotransmitter receptor transport. The clinical similarities between VPS35 -linked PD and sporadic PD suggest that defects observed in cellular and animal models with the D620N VPS35 mutation may provide valuable insights into sporadic disease. In this review, we highlight the current knowledge surrounding VPS35 and its role in retromer dysfunction in PD. We provide a critical discussion of the mechanisms implicated in VPS35 -mediated neurodegeneration in PD, as well as the interplay between VPS35 and other PD-linked gene products. This article is part of a discussion meeting issue 'Understanding the endo-lysosomal network in neurodegeneration'.
- Published
- 2024
- Full Text
- View/download PDF
8. Intracellular trafficking of HIV-1 Gag via Syntaxin 6-positive compartments/vesicles: Involvement in tumor necrosis factor secretion.
- Author
-
Tsurutani N, Momose F, Ogawa K, Sano K, and Morikawa Y
- Subjects
- Endosomes metabolism, Protein Transport genetics, Protein Binding, Protein Domains, HIV Infections metabolism, HIV Infections virology, Humans, Cell Line, Virus Replication genetics, HIV-1 genetics, HIV-1 metabolism, Qa-SNARE Proteins genetics, Qa-SNARE Proteins metabolism, Tumor Necrosis Factor-alpha metabolism, gag Gene Products, Human Immunodeficiency Virus genetics, gag Gene Products, Human Immunodeficiency Virus metabolism, Transport Vesicles metabolism
- Abstract
HIV-1 Gag protein is synthesized in the cytosol and is transported to the plasma membrane, where viral particle assembly and budding occur. Endosomes are alternative sites of Gag accumulation. However, the intracellular transport pathways and carriers for Gag have not been clarified. We show here that Syntaxin6 (Syx6), a soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) involved in membrane fusion in post-Golgi networks, is a molecule responsible for Gag trafficking and also for tumor necrosis factor-α (TNFα) secretion and that Gag and TNFα are cotransported via Syx6-positive compartments/vesicles. Confocal and live-cell imaging revealed that Gag colocalized and cotrafficked with Syx6, a fraction of which localizes in early and recycling endosomes. Syx6 knockdown reduced HIV-1 particle production, with Gag distributed diffusely throughout the cytoplasm. Coimmunoprecipitation and pulldown show that Gag binds to Syx6, but not its SNARE partners or their assembly complexes, suggesting that Gag preferentially binds free Syx6. The Gag matrix domain and the Syx6 SNARE domain are responsible for the interaction and cotrafficking. In immune cells, Syx6 knockdown/knockout similarly impaired HIV-1 production. Interestingly, HIV-1 infection facilitated TNFα secretion, and this enhancement did not occur in Syx6-depleted cells. Confocal and live-cell imaging revealed that TNFα and Gag partially colocalized and were cotransported via Syx6-positive compartments/vesicles. Biochemical analyses indicate that TNFα directly binds the C-terminal domain of Syx6. Altogether, our data provide evidence that both Gag and TNFα make use of Syx6-mediated trafficking machinery and suggest that Gag expression does not inhibit but rather facilitates TNFα secretion in HIV-1 infection., Competing Interests: Conflict of 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 Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
9. AP-1γ2 is an adaptor protein 1 variant required for endosome-to-Golgi trafficking of the mannose-6-P receptor (CI-MPR) and ATP7B copper transporter.
- Author
-
Tavares LA, Rodrigues RL, Santos da Costa C, Nascimento JA, Vargas de Carvalho J, Nogueira de Carvalho A, Mardones GA, and daSilva LLP
- Subjects
- Humans, HeLa Cells, Adaptor Protein Complex gamma Subunits metabolism, Endosomes metabolism, Protein Transport genetics, Receptor, IGF Type 2 genetics, Receptor, IGF Type 2 metabolism, trans-Golgi Network genetics, trans-Golgi Network metabolism, Copper-Transporting ATPases genetics, Copper-Transporting ATPases metabolism, Adaptor Protein Complex 1 genetics, Adaptor Protein Complex 1 metabolism
- Abstract
Selective retrograde transport from endosomes back to the trans-Golgi network (TGN) is important for maintaining protein homeostasis, recycling receptors, and returning molecules that were transported to the wrong compartments. Two important transmembrane proteins directed to this pathway are the Cation-Independent Mannose-6-phosphate receptor (CI-MPR) and the ATP7B copper transporter. Among CI-MPR functions is the delivery of acid hydrolases to lysosomes, while ATP7B facilitates the transport of cytosolic copper ions into organelles or the extracellular space. Precise subcellular localization of CI-MPR and ATP7B is essential for the proper functioning of these proteins. This study shows that both CI-MPR and ATP7B interact with a variant of the clathrin adaptor 1 (AP-1) complex that contains a specific isoform of the γ-adaptin subunit called γ2. Through synchronized anterograde trafficking and cell-surface uptake assays, we demonstrated that AP-1γ2 is dispensable for ATP7B and CI-MPR exit from the TGN while being critically required for ATP7B and CI-MPR retrieval from endosomes to the TGN. Moreover, AP-1γ2 depletion leads to the retention of endocytosed CI-MPR in endosomes enriched in retromer complex subunits. These data underscore the importance of AP-1γ2 as a key component in the sorting and trafficking machinery of CI-MPR and ATP7B, highlighting its essential role in the transport of proteins from endosomes., Competing Interests: Conflict of 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 Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
10. TMED2 promotes glioma tumorigenesis by being involved in EGFR recycling transport.
- Author
-
Sun C, Zhang Y, Wang Z, Chen J, Zhang J, and Gu Y
- Subjects
- Animals, Mice, Carcinogenesis genetics, Cell Line, Tumor, Cell Proliferation, Cell Transformation, Neoplastic, Signal Transduction, Humans, Membrane Proteins genetics, Membrane Proteins metabolism, Protein Transport genetics, ErbB Receptors metabolism, Glioma genetics, Glioma metabolism, Glioma pathology, Vesicular Transport Proteins genetics, Vesicular Transport Proteins metabolism
- Abstract
Aberrant epidermal growth factor receptor (EGFR) signaling is the core signaling commonly activated in glioma. The transmembrane emp24 protein transport domain protein 2 (TMED2) interacts with cargo proteins involved in protein sorting and transport between endoplasmic reticulum (ER) and Golgi apparatus. In this study, we found the correlation between TMED2 with glioma progression and EGFR signaling through database analysis. Moreover, we demonstrated that TMED2 is essential for glioma cell proliferation, migration, and invasion at the cellular levels, as well as tumor formation in mouse models, underscoring its significance in the pathobiology of gliomas. Mechanistically, TMED2 was found to enhance EGFR-AKT signaling by facilitating EGFR recycling, thereby providing the initial evidence of TMED2's involvement in the membrane protein recycling process. In summary, our findings shed light on the roles and underlying mechanisms of TMED2 in the regulation of glioma tumorigenesis and EGFR signaling, suggesting that targeting TMED2 could emerge as a promising therapeutic strategy for gliomas and other tumors associated with aberrant EGFR signaling., 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 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
11. The P2Y 2 receptor mediates terminal adipocyte differentiation and insulin resistance: Evidence for a dual G-protein coupling mode.
- Author
-
Qian S, Shi Y, Senfeld J, Peng Q, and Shen J
- Subjects
- Animals, Humans, Mice, Adipocytes cytology, Adipocytes metabolism, GTP-Binding Proteins metabolism, Proto-Oncogene Proteins c-akt genetics, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction genetics, Cells, Cultured, Mice, Inbred C57BL, Up-Regulation, Glucose Transporter Type 4 metabolism, Protein Transport genetics, Lipolysis genetics, Insulin Resistance genetics, Receptors, Purinergic P2Y2 genetics, Receptors, Purinergic P2Y2 metabolism, Adipogenesis genetics
- Abstract
Several P2Y nucleotide receptors have been shown to be involved in the early stage of adipocyte differentiation in vitro and insulin resistance in obese mice; however, the exact receptor subtype(s) and its underlying molecular mechanism in relevant human cells are unclear. Here, using human primary visceral preadipocytes as a model, we found that during preadipocyte-to-mature adipocyte differentiation, the P2Y
2 nucleotide receptor (P2Y2 R) was the most upregulated subtype among the eight known P2Y receptors and the only one further dramatically upregulated after inflammatory TNFα treatment. Functional studies indicated that the P2Y2 R induced intracellular Ca2+ , ERK1/2, and JNK signaling but not the p38 pathway. In addition, stimulation of the P2Y2 R suppressed basal and insulin-induced phosphorylation of AKT, accompanied by decreased GLUT4 membrane translocation and glucose uptake in mature adipocytes, suggesting a role of P2Y2 R in insulin resistance. Mechanistically, we found that activation of P2Y2 R did not increase lipolysis but suppressed PIP3 generation. Interestingly, activation of P2Y2 R triggered Gi -protein coupling, and pertussis toxin pretreatment largely inhibited P2Y2 R-mediated ERK1/2 signaling and cAMP suppression. Further, treatment of the cells with AR-C 118925XX, a selective P2Y2 R antagonist, significantly inhibited adipogenesis, and P2Y2 R knockout decreased mouse body weight gain with smaller eWAT mass infiltrated with fewer macrophages as compared to WT mice in response to a Western diet. Thus, we revealed that terminal adipocyte differentiation and inflammation selectively upregulate P2Y2 R expression and that P2Y2 R mediates insulin resistance by suppressing the AKT signaling pathway, highlighting P2Y2 R as a potential new drug target to combat obesity and type-2 diabetes., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
12. Endogenous Tagging of Ciliary Genes in Human RPE1 Cells for Live-Cell Imaging.
- Author
-
Kuhns S, Juhl AD, Anvarian Z, Wüstner D, Pedersen LB, and Andersen JS
- Subjects
- Humans, Protein Transport genetics, Biological Transport, Cilia metabolism, Proteins metabolism
- Abstract
CRISPR-mediated endogenous tagging of genes provides unique possibilities to explore the function and dynamic subcellular localization of proteins in living cells. Here, we describe experimental strategies for endogenous PCR-tagging of ciliary genes in human RPE1 cells and how image acquisition and analysis of the expressed fluorescently tagged proteins can be utilized to study the dynamic ciliary processes of intraflagellar transport and vesicular trafficking., (© 2024. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
13. POMC Neuron BBSome Regulation of Body Weight is Independent of its Ciliary Function.
- Author
-
Guo DF, Williams PA, Laule C, Seaby C, Zhang Q, Sheffield VC, and Rahmouni K
- Subjects
- Humans, Glucose metabolism, Microtubule-Associated Proteins genetics, Neurons metabolism, Protein Transport genetics, Serotonin metabolism, Animals, Bardet-Biedl Syndrome, Body Weight, Cilia genetics, Pro-Opiomelanocortin genetics
- Abstract
The BBSome, a complex of several Bardet-Biedl syndrome (BBS) proteins including BBS1, has emerged as a critical regulator of energy homeostasis. Although the BBSome is best known for its involvement in cilia trafficking, through a process that involve BBS3, it also regulates the localization of cell membrane receptors underlying metabolic regulation. Here, we show that inducible Bbs1 gene deletion selectively in proopiomelanocortin (POMC) neurons cause a gradual increase in body weight, which was associated with higher fat mass. In contrast, inducible deletion of Bbs3 gene in POMC neurons failed to affect body weight and adiposity. Interestingly, loss of BBS1 in POMC neurons led to glucose intolerance and insulin insensitivity, whereas BBS3 deficiency in these neurons is associated with slight impairment in glucose handling, but normal insulin sensitivity. BBS1 deficiency altered the plasma membrane localization of serotonin 5-HT2C receptor (5-HT
2C R) and ciliary trafficking of neuropeptide Y2 receptor (NPY2 R).In contrast, BBS3 deficiency, which disrupted the ciliary localization of the BBSome, did not interfere with plasma membrane expression of 5-HT2C R, but reduced the trafficking of NPY2 R to cilia. We also show that deficiency in BBS1, but not BBS3, alters mitochondria dynamics and decreased total and phosphorylated levels of dynamin-like protein 1 (DRP1) protein. Importantly, rescuing DRP1 activity restored mitochondria dynamics and localization of 5-HT2C R and NPY2 R in BBS1-deficient cells. The contrasting effects on energy and glucose homeostasis evoked by POMC neuron deletion of BBS1 versus BBS3 indicate that BBSome regulation of metabolism is not related to its ciliary function in these neurons., Competing Interests: The authors have no conflict of interest relevant to this study., (© The Author(s) 2023. Published by Oxford University Press on behalf of American Physiological Society.)- Published
- 2023
- Full Text
- View/download PDF
14. CD8 + T cells reduce neuroretina inflammation in mouse by regulating autoreactive Th1 and Th17 cells through IFN-γ.
- Author
-
Wu S, Zhang X, Hu C, Zhong Y, Chen J, and Chong WP
- Subjects
- Male, Female, Animals, Mice, Mice, Inbred C57BL, Lymphocyte Activation, Th1 Cells immunology, Th17 Cells immunology, Cell Polarity immunology, Interleukin-10 immunology, Interferon-beta pharmacology, Receptors, CXCR3 genetics, Receptors, CXCR3 immunology, Protein Transport genetics, Spleen immunology, CD8-Positive T-Lymphocytes drug effects, CD8-Positive T-Lymphocytes immunology, Retinitis immunology, Interferon-gamma immunology
- Abstract
Various regulatory CD8
+ T-cell subsets have been proposed for immune tolerance and have been implicated in controlling autoimmune diseases. However, their phenotypic identities and suppression mechanisms are not yet understood. This study found that coculture of T-cell receptor (TCR)- or interferon (IFN)-β-activated CD8+ T cells significantly suppressed the cytokine production of Th1 and Th17 cells. By experimenting with the experimental autoimmune uveitis (EAU), we found that adoptive transfer of TCR or IFN-β-activated CD8+ T cells significantly lessened disease development in an IFN-γ-dependent manner with a decreased uveitogenic Th1 and Th17 response. Interestingly, after adoptive transfer into the EAU mice, the IFN-γ+ CD8+ T cells were recruited more efficiently into the secondary lymphoid organs during the disease-priming phase. This recruitment depends on the IFN-γ-inducible chemokine receptor CXCR3; knocking out CXCR3 abolishes the protective effect of CD8+ T cells in EAU. In conclusion, we identified the critical role of IFN-γ for CD8+ T cells to inhibit Th1 and Th17 responses and ameliorate EAU. CXCR3 is necessary to recruit IFN-γ+ CD8+ T cells to the secondary lymphoid organ for the regulation of autoreactive Th1 and Th17 cells., (© 2023 The Authors. European Journal of Immunology published by Wiley-VCH GmbH.)- Published
- 2023
- Full Text
- View/download PDF
15. The plant-unique protein DRIF1 coordinates with sorting nexin 1 to regulate membrane protein homeostasis.
- Author
-
Zhu Y, Zhao Q, Cao W, Huang S, Ji C, Zhang W, Trujillo M, Shen J, and Jiang L
- Subjects
- Sorting Nexins genetics, Sorting Nexins metabolism, Plant Proteins metabolism, Endosomal Sorting Complexes Required for Transport metabolism, Proteostasis, Protein Transport genetics, Plants metabolism, Vesicular Transport Proteins genetics, Vesicular Transport Proteins metabolism, Arabidopsis metabolism, Arabidopsis Proteins metabolism
- Abstract
Membrane protein homeostasis is fine-tuned by the cellular pathways for vacuolar degradation and recycling, which ultimately facilitate plant growth and cell-environment interactions. The endosomal sorting complex required for transport (ESCRT) machinery plays important roles in regulating intraluminal vesicle (ILV) formation and membrane protein sorting to vacuoles. We previously showed that the plant-specific ESCRT component FYVE DOMAIN PROTEIN REQUIRED FOR ENDOSOMAL SORTING1 (FREE1) performs multiple functions in plants, although the underlying mechanisms remain elusive. In this study, we performed a suppressor screen of the FREE1-RNAi mutant and identified and characterized 2 suppressor of free1 (sof) mutants in Arabidopsis (Arabidopsis thaliana). These mutants, sof10 and sof641, result in a premature stop codon or a missense mutation in AT5G10370, respectively. This gene was named DEAH and RING domain-containing protein as FREE1 suppressor 1 (DRIF1). DRIF1 has a homologous gene, DRIF2, in the Arabidopsis genome with 95% identity to DRIF1. The embryos of drif1 drif2 mutants arrested at the globular stage and formed enlarged multivesicular bodies (MVBs) with an increased number of ILVs. DRIF1 is a membrane-associated protein that coordinates with retromer component sorting nexin 1 to regulate PIN-FORMED2 recycling to the plasma membrane. Altogether, our data demonstrate that DRIF1 is a unique retromer interactor that orchestrates FREE1-mediated ILV formation of MVBs and vacuolar sorting of membrane proteins for degradation in plants., Competing Interests: Conflict of interest statement. The authors declare no conflict of interest., (© American Society of Plant Biologists 2023. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
16. Intralumenal docking of connexin 36 channels in the ER isolates mistrafficked protein.
- Author
-
Tetenborg S, Liss V, Breitsprecher L, Timonina K, Kotova A, Acevedo Harnecker AJ, Yuan C, Shihabeddin E, Ariakia F, Qin G, Chengzhi C, Dedek K, Zoidl G, Hensel M, and O'Brien J
- Subjects
- Humans, HEK293 Cells, Protein Domains, Amino Acid Motifs, Electrical Synapses physiology, Mutation, Protein Transport genetics, Synaptic Vesicles pathology, Synaptic Vesicles ultrastructure, Microscopy, Electron, Scanning, Gap Junction delta-2 Protein, Connexins genetics, Connexins metabolism, Endoplasmic Reticulum metabolism, Gap Junctions metabolism
- Abstract
The intracellular domains of connexins are essential for the assembly of gap junctions. For connexin 36 (Cx36), the major neuronal connexin, it has been shown that a dysfunctional PDZ-binding motif interferes with electrical synapse formation. However, it is still unknown how this motif coordinates the transport of Cx36. In the present study, we characterize a phenotype of Cx36 mutants that lack a functional PDZ-binding motif using HEK293T cells as an expression system. We provide evidence that an intact PDZ-binding motif is critical for proper endoplasmic reticulum (ER) export of Cx36. Removing the PDZ-binding motif of Cx36 results in ER retention and the formation of multimembrane vesicles containing gap junction-like connexin aggregates. Using a combination of site-directed mutagenesis and electron micrographs, we reveal that these vesicles consist of Cx36 channels that docked prematurely in the ER. Our data suggest a model in which ER-retained Cx36 channels reshape the ER membrane into concentric whorls that are released into the cytoplasm., Competing Interests: Conflicts of interests The authors declare that they have no conflicts of interests with the contents of this article., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
17. ESCRT-dependent control of craniofacial morphogenesis with concomitant perturbation of NOTCH signaling.
- Author
-
Hermosilla Aguayo V, Martin P, Tian N, Zheng J, Aho R, Losa M, and Selleri L
- Subjects
- Animals, Humans, Mice, Protein Transport genetics, Signal Transduction, Morphogenesis, Endosomes metabolism, Endosomal Sorting Complexes Required for Transport genetics, Endosomal Sorting Complexes Required for Transport metabolism, Carrier Proteins metabolism
- Abstract
Craniofacial development is orchestrated by transcription factor-driven regulatory networks, epigenetic modifications, and signaling pathways. Signaling molecules and their receptors rely on endo-lysosomal trafficking to prevent accumulation on the plasma membrane. ESCRT (Endosomal Sorting Complexes Required for Transport) machinery is recruited to endosomal membranes enabling degradation of such endosomal cargoes. Studies in vitro and in invertebrate models established the requirements of the ESCRT machinery in membrane remodeling, endosomal trafficking, and lysosomal degradation of activated membrane receptors. However, investigations during vertebrate development have been scarce. By ENU-induced mutagenesis, we isolated a mouse line, Vps25
ENU/ENU , carrying a hypomorphic allele of the ESCRT-II component Vps25, with craniofacial anomalies resembling features of human congenital syndromes. Here, we assessed the spatiotemporal dynamics of Vps25 and additional ESCRT-encoding genes during murine development. We show that these genes are ubiquitously expressed although enriched in discrete domains of the craniofacial complex, heart, and limbs. ESCRT-encoding genes, including Vps25, are expressed in both cranial neural crest-derived mesenchyme and epithelium. Unlike constitutive ESCRT mutants, Vps25ENU/ENU embryos display late lethality. They exhibit hypoplastic lower jaw, stunted snout, dysmorphic ear pinnae, and secondary palate clefting. Thus, we provide the first evidence for critical roles of ESCRT-II in craniofacial morphogenesis and report perturbation of NOTCH signaling in craniofacial domains of Vps25ENU/ENU embryos. Given the known roles of NOTCH signaling in the developing cranium, and notably the lower jaw, we propose that the NOTCH pathway partly mediates the craniofacial defects of Vps25ENU/ENU mouse embryos., (Copyright © 2023. Published by Elsevier Inc.)- Published
- 2023
- Full Text
- View/download PDF
18. Comparative proximity biotinylation implicates the small GTPase RAB18 in sterol mobilization and biosynthesis.
- Author
-
Kiss RS, Chicoine J, Khalil Y, Sladek R, Chen H, Pisaturo A, Martin C, Dale JD, Brudenell TA, Kamath A, Kyei-Boahen J, Hafiane A, Daliah G, Alecki C, Hopes TS, Heier M, Aligianis IA, Lebrun JJ, Aspden J, Paci E, Kerksiek A, Lütjohann D, Clayton P, Wills JC, von Kriegsheim A, Nilsson T, Sheridan E, and Handley MT
- Subjects
- Humans, Cells, Cultured, Cholesterol biosynthesis, Cholesterol metabolism, Gene Knockdown Techniques, Guanine Nucleotide Exchange Factors genetics, Guanine Nucleotide Exchange Factors metabolism, HeLa Cells, Protein Transport genetics, rab3 GTP-Binding Proteins metabolism, Calcium Channels genetics, Calcium Channels metabolism, Biotinylation, rab GTP-Binding Proteins genetics, rab GTP-Binding Proteins metabolism, Sterols biosynthesis, Sterols metabolism
- Abstract
Loss of functional RAB18 causes the autosomal recessive condition Warburg Micro syndrome. To better understand this disease, we used proximity biotinylation to generate an inventory of potential RAB18 effectors. A restricted set of 28 RAB18 interactions were dependent on the binary RAB3GAP1-RAB3GAP2 RAB18-guanine nucleotide exchange factor complex. Twelve of these 28 interactions are supported by prior reports, and we have directly validated novel interactions with SEC22A, TMCO4, and INPP5B. Consistent with a role for RAB18 in regulating membrane contact sites, interactors included groups of microtubule/membrane-remodeling proteins, membrane-tethering and docking proteins, and lipid-modifying/transporting proteins. Two of the putative interactors, EBP and OSBPL2/ORP2, have sterol substrates. EBP is a Δ8-Δ7 sterol isomerase, and ORP2 is a lipid transport protein. This prompted us to investigate a role for RAB18 in cholesterol biosynthesis. We found that the cholesterol precursor and EBP-product lathosterol accumulates in both RAB18-null HeLa cells and RAB3GAP1-null fibroblasts derived from an affected individual. Furthermore, de novo cholesterol biosynthesis is impaired in cells in which RAB18 is absent or dysregulated or in which ORP2 expression is disrupted. Our data demonstrate that guanine nucleotide exchange factor-dependent Rab interactions are highly amenable to interrogation by proximity biotinylation and may suggest that Micro syndrome is a cholesterol biosynthesis disorder., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
19. A multiple-oscillator mechanism underlies antigen-induced Ca 2+ oscillations in Jurkat T-cells.
- Author
-
Benson JC, Romito O, Abdelnaby AE, Xin P, Pathak T, Weir SE, Kirk V, Castaneda F, Yoast RE, Emrich SM, Tang PW, Yule DI, Hempel N, Potier-Cartereau M, Sneyd J, and Trebak M
- Subjects
- Humans, Calcium metabolism, Jurkat Cells, Stromal Interaction Molecule 1 genetics, Stromal Interaction Molecule 1 metabolism, Stromal Interaction Molecule 2 genetics, Stromal Interaction Molecule 2 metabolism, Gene Knockout Techniques, Models, Biological, Protein Isoforms, Protein Transport genetics, Cell Proliferation genetics, Cell Survival genetics, Calcium Release Activated Calcium Channels genetics, Calcium Release Activated Calcium Channels metabolism, Calcium Signaling genetics
- Abstract
T-cell receptor stimulation triggers cytosolic Ca
2+ signaling by inositol-1,4,5-trisphosphate (IP3 )-mediated Ca2+ release from the endoplasmic reticulum (ER) and Ca2+ entry through Ca2+ release-activated Ca2+ (CRAC) channels gated by ER-located stromal-interacting molecules (STIM1/2). Physiologically, cytosolic Ca2+ signaling manifests as regenerative Ca2+ oscillations, which are critical for nuclear factor of activated T-cells-mediated transcription. In most cells, Ca2+ oscillations are thought to originate from IP3 receptor-mediated Ca2+ release, with CRAC channels indirectly sustaining them through ER refilling. Here, experimental and computational evidence support a multiple-oscillator mechanism in Jurkat T-cells whereby both IP3 receptor and CRAC channel activities oscillate and directly fuel antigen-evoked Ca2+ oscillations, with the CRAC channel being the major contributor. KO of either STIM1 or STIM2 significantly reduces CRAC channel activity. As such, STIM1 and STIM2 synergize for optimal Ca2+ oscillations and activation of nuclear factor of activated T-cells 1 and are essential for ER refilling. The loss of both STIM proteins abrogates CRAC channel activity, drastically reduces ER Ca2+ content, severely hampers cell proliferation and enhances cell death. These results clarify the mechanism and the contribution of STIM proteins to Ca2+ oscillations in T-cells., Competing Interests: Conflict of interest Mohamed Trebak is a consultant for Seeker Biologics Inc, and is a member of the editorial board of the J. Biol. Chem. The remaining authors declare no competing interests., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
20. Sialylation of EGFR by ST6GAL1 induces receptor activation and modulates trafficking dynamics.
- Author
-
Ankenbauer KE, Rao TC, Mattheyses AL, and Bellis SL
- Subjects
- Humans, Cell Line, Tumor, Ovarian Neoplasms physiopathology, Signal Transduction, Protein Transport genetics, Protein Binding, beta-D-Galactoside alpha 2-6-Sialyltransferase genetics, beta-D-Galactoside alpha 2-6-Sialyltransferase metabolism, ErbB Receptors genetics, ErbB Receptors metabolism
- Abstract
Aberrant glycosylation is a hallmark of a cancer cell. One prevalent alteration is an enrichment in α2,6-linked sialylation of N-glycosylated proteins, a modification directed by the ST6GAL1 sialyltransferase. ST6GAL1 is upregulated in many malignancies including ovarian cancer. Prior studies have shown that the addition of α2,6 sialic acid to the epidermal growth factor receptor (EGFR) activates this receptor, although the mechanism was largely unknown. To investigate the role of ST6GAL1 in EGFR activation, ST6GAL1 was overexpressed in the OV4 ovarian cancer line, which lacks endogenous ST6GAL1, or knocked-down in the OVCAR-3 and OVCAR-5 ovarian cancer lines, which have robust ST6GAL1 expression. Cells with high expression of ST6GAL1 displayed increased activation of EGFR and its downstream signaling targets, AKT and NFκB. Using biochemical and microscopy approaches, including total internal reflection fluorescence microscopy, we determined that the α2,6 sialylation of EGFR promoted its dimerization and higher order oligomerization. Additionally, ST6GAL1 activity was found to modulate EGFR trafficking dynamics following EGF-induced receptor activation. Specifically, EGFR sialylation enhanced receptor recycling to the cell surface following activation while simultaneously inhibiting lysosomal degradation. 3D widefield deconvolution microscopy confirmed that in cells with high ST6GAL1 expression, EGFR exhibited greater colocalization with Rab11 recycling endosomes and reduced colocalization with LAMP1-positive lysosomes. Collectively, our findings highlight a novel mechanism by which α2,6 sialylation promotes EGFR signaling by facilitating receptor oligomerization and recycling., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
21. Alternative splicing regulates adaptor protein binding, trafficking, and activity of the Vps10p domain receptor SorCS2 in neuronal development.
- Author
-
Skeldal S, Voss LF, Lende J, Pedersen SB, Mølgaard S, Kaas M, Demange P, Bentsen AH, Fuglsang M, Sander MR, Buttenschøn H, Gustafsen C, Madsen P, and Glerup S
- Subjects
- Humans, Adaptor Proteins, Signal Transducing metabolism, Brain-Derived Neurotrophic Factor genetics, Brain-Derived Neurotrophic Factor metabolism, Dyneins metabolism, Kinesins metabolism, Protein Binding, Protein Isoforms metabolism, Receptor, trkB metabolism, Protein Processing, Post-Translational, Protein Transport genetics, Alternative Splicing physiology, Receptors, Cell Surface metabolism, Central Nervous System growth & development
- Abstract
The Vps10p domain receptor SorCS2 is crucial for the development and function of the nervous system and essential for brain-derived neurotrophic factor (BDNF)-induced changes in neuronal morphology and plasticity. SorCS2 regulates the subcellular trafficking of the BDNF signaling receptor TrkB as well as selected neurotransmitter receptors in a manner that is dependent on the SorCS2 intracellular domain (ICD). However, the cellular machinery and adaptor protein (AP) interactions that regulate receptor trafficking via the SorCS2 ICD are unknown. We here identify four splice variants of human SorCS2 differing in the insertion of an acidic cluster motif and/or a serine residue within the ICD. We show that each variant undergoes posttranslational proteolytic processing into a one- or two-chain receptor, giving rise to eight protein isoforms, the expression of which differs between neuronal and nonneuronal tissues and is affected by cellular stressors. We found that the only variants without the serine were able to rescue BDNF-induced branching of SorCS2 knockout hippocampal neurons, while variants without the acidic cluster showed increased interactions with clathrin-associated APs AP-1, AP-2, and AP-3. Using yeast two-hybrid screens, we further discovered that all variants bound dynein light chain Tctex-type 3; however, only variants with an acidic cluster motif bound kinesin light chain 1. Accordingly, splice variants showed markedly different trafficking properties and localized to different subcellular compartments. Taken together, our findings demonstrate the existence of eight functional SorCS2 isoforms with differential capacity for interactions with cytosolic ligands dynein light chain Tctex-type 3 and kinesin light chain 1, which potentially allows cell-type specific SorCS2 trafficking and BDNF signaling., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
22. RABL2 promotes the outward transition zone passage of signaling proteins in cilia via ARL3.
- Author
-
Zhang RK, Sun WY, Liu YX, Zhang EY, and Fan ZC
- Subjects
- Humans, ADP-Ribosylation Factors genetics, ADP-Ribosylation Factors metabolism, GTP Phosphohydrolases metabolism, Guanosine Triphosphate metabolism, Membrane Proteins metabolism, Protein Transport genetics, rab GTP-Binding Proteins metabolism, Chlamydomonas, Cilia metabolism, Hedgehog Proteins metabolism
- Abstract
Certain transmembrane and membrane-tethered signaling proteins export from cilia as BBSome cargoes via the outward BBSome transition zone (TZ) diffusion pathway, indispensable for maintaining their ciliary dynamics to enable cells to sense and transduce extracellular stimuli inside the cell. Murine Rab-like 2 (Rabl2) GTPase resembles Chlamydomonas Arf-like 3 (ARL3) GTPase in promoting outward TZ passage of the signaling protein cargo-laden BBSome. During this process, ARL3 binds to and recruits the retrograde IFT train-dissociated BBSome as its effector to diffuse through the TZ for ciliary retrieval, while how RABL2 and ARL3 cross talk in this event remains uncertain. Here, we report that Chlamydomonas RABL2 in a GTP-bound form (RABL2
GTP ) cycles through cilia via IFT as an IFT-B1 cargo, dissociates from retrograde IFT trains at a ciliary region right above the TZ, and converts to RABL2GDP for activating ARL3GDP as an ARL3 guanine nucleotide exchange factor. This confers ARL3GTP to detach from the ciliary membrane and become available for binding and recruiting the phospholipase D (PLD)-laden BBSome, autonomous of retrograde IFT association, to diffuse through the TZ for ciliary retrieval. Afterward, RABL2GDP exits cilia by being bound to the ARL3GTP /BBSome entity as a BBSome cargo. Our data identify ciliary signaling proteins exported from cilia via the RABL2-ARL3 cascade-mediated outward BBSome TZ diffusion pathway. According to this model, hedgehog signaling defect-induced Bardet-Biedl syndrome caused by RABL2 mutations in humans could be well explained in a mutation-specific manner, providing us with a mechanistic understanding behind the outward BBSome TZ passage required for proper ciliary signaling.- Published
- 2023
- Full Text
- View/download PDF
23. ESCRT-III component OsSNF7.2 modulates leaf rolling by trafficking and endosomal degradation of auxin biosynthetic enzyme OsYUC8 in rice.
- Author
-
Zhou L, Chen S, Cai M, Cui S, Ren Y, Zhang X, Liu T, Zhou C, Jin X, Zhang L, Wu M, Zhang S, Cheng Z, Zhang X, Lei C, Lin Q, Guo X, Wang J, Zhao Z, Jiang L, Zhu S, and Wan J
- Subjects
- Plant Proteins genetics, Plant Proteins metabolism, Endosomes metabolism, Plant Leaves metabolism, Protein Transport genetics, Endosomal Sorting Complexes Required for Transport genetics, Endosomal Sorting Complexes Required for Transport metabolism, Oryza metabolism
- Abstract
The endosomal sorting complex required for transport (ESCRT) is highly conserved in eukaryotic cells and plays an essential role in the biogenesis of multivesicular bodies and cargo degradation to the plant vacuole or lysosomes. Although ESCRT components affect a variety of plant growth and development processes, their impact on leaf development is rarely reported. Here, we found that OsSNF7.2, an ESCRT-III component, controls leaf rolling in rice (Oryza sativa). The Ossnf7.2 mutant rolled leaf 17 (rl17) has adaxially rolled leaves due to the decreased number and size of the bulliform cells. OsSNF7.2 is expressed ubiquitously in all tissues, and its protein is localized in the endosomal compartments. OsSNF7.2 homologs, including OsSNF7, OsSNF7.3, and OsSNF7.4, can physically interact with OsSNF7.2, but their single mutation did not result in leaf rolling. Other ESCRT complex subunits, namely OsVPS20, OsVPS24, and OsBRO1, also interact with OsSNF7.2. Further assays revealed that OsSNF7.2 interacts with OsYUC8 and aids its vacuolar degradation. Both Osyuc8 and rl17 Osyuc8 showed rolled leaves, indicating that OsYUC8 and OsSNF7.2 function in the same pathway, conferring leaf development. This study reveals a new biological function for the ESCRT-III components, and provides new insights into the molecular mechanisms underlying leaf rolling., (© 2023 Institute of Botany, Chinese Academy of Sciences.)
- Published
- 2023
- Full Text
- View/download PDF
24. Defective mitochondrial import as a challenge for cellular protein homeostasis.
- Author
-
Maruszczak KK, Ayyamperumal S, and Chacinska A
- Subjects
- Mitochondrial Membranes metabolism, Mitochondrial Proteins genetics, Mitochondrial Proteins metabolism, Carrier Proteins metabolism, Protein Transport genetics, Homeostasis, Proteostasis, Mitochondria metabolism
- Abstract
Mitochondria are organelles indispensable for the correct functioning of eukaryotic cells. Their significance for cellular homeostasis is manifested by the existence of complex quality control pathways that monitor organellar fitness. Mitochondrial biogenesis relies on the efficient import of mitochondrial precursor proteins, a large majority of which are encoded by nuclear DNA and synthesized in the cytosol. This creates a demand for highly specialized import routes that comprise cytosolic factors and organellar translocases. The passage of newly encoded mitochondrial precursor proteins through the cytosol to the translocase of the outer mitochondrial membrane (TOM) is under tight surveillance. As a result of mitochondrial import defects, mitochondrial precursor proteins accumulate in the cytosol or clog the TOM complex, which in turn stimulates cellular stress responses to minimize the consequences of these challenges. These responses are critical for maintaining protein homeostasis under conditions of mitochondrial stress. The present review summarizes recent advances in the field of mitochondrial protein import quality control and discusses the role of this quality control within the network of cellular mechanisms that maintain the cellular homeostasis of proteins., (© 2023 Federation of European Biochemical Societies.)
- Published
- 2023
- Full Text
- View/download PDF
25. PKA mediates modality-specific modulation of the mechanically gated ion channel PIEZO2.
- Author
-
Schaefer I, Verkest C, Vespermann L, Mair T, Voß H, Zeitzschel N, and Lechner SG
- Subjects
- Humans, Pain physiopathology, Protein Domains, Protein Transport genetics, Ion Channels genetics, Ion Channels metabolism, Mechanotransduction, Cellular genetics, Cyclic AMP-Dependent Protein Kinases metabolism
- Abstract
PKA is a downstream effector of many inflammatory mediators that induce pain hypersensitivity by increasing the mechanosensitivity of nociceptive sensory afferent. Here, we examine the molecular mechanism underlying PKA-dependent modulation of the mechanically activated ion channel PIEZO2, which confers mechanosensitivity to many nociceptors. Using phosphorylation site prediction algorithms, we identified multiple putative and highly conserved PKA phosphorylation sites located on intracellular intrinsically disordered regions of PIEZO2. Site-directed mutagenesis and patch-clamp recordings showed that substitution of one or multiple putative PKA sites within a single intracellular domain does not alter PKA-induced PIEZO2 sensitization, whereas mutation of a combination of nine putative sites located on four different intracellular regions completely abolishes PKA-dependent PIEZO2 modulation, though it remains unclear whether all or just some of these nine sites are required. By demonstrating that PIEZO1 is not modulated by PKA, our data also reveal a previously unrecognized functional difference between PIEZO1 and PIEZO2. Moreover, by demonstrating that PKA only modulates PIEZO2 currents evoked by focal mechanical indentation of the cell, but not currents evoked by pressure-induced membrane stretch, we provide evidence suggesting that PIEZO2 is a polymodal mechanosensor that engages different protein domains for detecting different types of mechanical stimuli., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
26. The ribosomal S6 kinase 2 (RSK2)-SPRED2 complex regulates the phosphorylation of RSK substrates and MAPK signaling.
- Author
-
Lopez J, Bonsor DA, Sale MJ, Urisman A, Mehalko JL, Cabanski-Dunning M, Castel P, Simanshu DK, and McCormick F
- Subjects
- Phosphorylation, Humans, Cell Line, Protein Domains, Gene Knockdown Techniques, Protein Transport genetics, Protein Binding, Protein Structure, Tertiary, Models, Molecular, Neurofibromin 1 metabolism, Mitogen-Activated Protein Kinases metabolism, Ribosomal Protein S6 Kinases, 90-kDa chemistry, Ribosomal Protein S6 Kinases, 90-kDa genetics, Ribosomal Protein S6 Kinases, 90-kDa metabolism, Signal Transduction genetics, Repressor Proteins chemistry, Repressor Proteins metabolism
- Abstract
Sprouty-related EVH-1 domain-containing (SPRED) proteins are a family of proteins that negatively regulate the RAS-Mitogen-Activated Protein Kinase (MAPK) pathway, which is involved in the regulation of the mitogenic response and cell proliferation. However, the mechanism by which these proteins affect RAS-MAPK signaling has not been elucidated. Patients with mutations in SPRED give rise to unique disease phenotypes; thus, we hypothesized that distinct interactions across SPRED proteins may account for alternative nodes of regulation. To characterize the SPRED interactome and evaluate how members of the SPRED family function through unique binding partners, we performed affinity purification mass spectrometry. We identified 90-kDa ribosomal S6 kinase 2 (RSK2) as a specific interactor of SPRED2 but not SPRED1 or SPRED3. We identified that the N-terminal kinase domain of RSK2 mediates the interaction between amino acids 123 to 201 of SPRED2. Using X-ray crystallography, we determined the structure of the SPRED2-RSK2 complex and identified the SPRED2 motif, F145A, as critical for interaction. We found that the formation of this interaction is regulated by MAPK signaling events. We also find that this interaction between SPRED2 and RSK2 has functional consequences, whereby the knockdown of SPRED2 resulted in increased phosphorylation of RSK substrates, YB1 and CREB. Furthermore, SPRED2 knockdown hindered phospho-RSK membrane and nuclear subcellular localization. We report that disruption of the SPRED2-RSK complex has effects on RAS-MAPK signaling dynamics. Our analysis reveals that members of the SPRED family have unique protein binding partners and describes the molecular and functional determinants of SPRED2-RSK2 complex dynamics., Competing Interests: Conflict of interest P. C is a founder and advisory board of Venthera, Inc. The other authors declare no competing interests. F. M. is a consultant for the following companies: Amgen; Daiichi Ltd, Frontier Medicines, Exuma Biotech, Ideaya Biosciences, Kura Oncology, Leidos Biomedical Research, Inc, PellePharm, Pfizer Inc, PMV Pharma and Quanta Therapeutics. F. M. is a consultant and co-founder for the following companies (with ownership interest including stock options): BridgeBio; DNAtrix Inc; Olema Pharmaceuticals, Inc; and Quartz. F. M. is the scientific director of the National Cancer Institute (NCI) RAS Initiative at the Frederick National Laboratory for Cancer Research/Leidos Biomedical Research, Inc. F. M. has been a recipient of research grants from Daiichi Sankyo, Gilead Sciences and has a current grant from Boehringer-Ingelheim., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
27. The adapter protein FADD provides an alternate pathway for entry into the cell cycle by regulating APC/C-Cdh1 E3 ubiquitin ligase activity.
- Author
-
Awadia S, Sitto M, Ram S, Ji W, Liu Y, Damani R, Ray D, Lawrence TS, Galban CJ, Cappell SD, and Rehemtulla A
- Subjects
- Humans, Adaptor Proteins, Signal Transducing metabolism, Anaphase-Promoting Complex-Cyclosome metabolism, Cell Cycle genetics, Cell Division, Gene Expression, HEK293 Cells, Mutation, Protein Domains, Protein Transport genetics, Cell Cycle Proteins metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism
- Abstract
The E3 ubiquitin ligase APC/C-Cdh1 maintains the G0/G1 state, and its inactivation is required for cell cycle entry. We reveal a novel role for Fas-associated protein with death domain (FADD) in the cell cycle through its function as an inhibitor of APC/C-Cdh1. Using real-time, single-cell imaging of live cells combined with biochemical analysis, we demonstrate that APC/C-Cdh1 hyperactivity in FADD-deficient cells leads to a G1 arrest despite persistent mitogenic signaling through oncogenic EGFR/KRAS. We further show that FADD
WT interacts with Cdh1, while a mutant lacking a consensus KEN-box motif (FADDKEN ) fails to interact with Cdh1 and results in a G1 arrest due to its inability to inhibit APC/C-Cdh1. Additionally, enhanced expression of FADDWT but not FADDKEN , in cells arrested in G1 upon CDK4/6 inhibition, leads to APC/C-Cdh1 inactivation and entry into the cell cycle in the absence of retinoblastoma protein phosphorylation. FADD's function in the cell cycle requires its phosphorylation by CK1α at Ser-194 which promotes its nuclear translocation. Overall, FADD provides a CDK4/6-Rb-E2F-independent "bypass" mechanism for cell cycle entry and thus a therapeutic opportunity for CDK4/6 inhibitor resistance., Competing Interests: Conflict of interests The authors declare that no actual, potential, or perceived conflict of interest or competing interests exists in relation to this research., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
28. Conserved NIMA kinases regulate multiple steps of endocytic trafficking.
- Author
-
Joseph BB, Naslavsky N, Binti S, Conquest S, Robison L, Bai G, Homer RO, Grant BD, Caplan S, and Fay DS
- Subjects
- Animals, Humans, Endocytosis genetics, Endosomes genetics, Endosomes metabolism, NIMA-Related Kinases genetics, NIMA-Related Kinases metabolism, Clathrin genetics, Clathrin metabolism, Bone Morphogenetic Protein Receptors metabolism, Protein Transport genetics, Caenorhabditis elegans metabolism, Caenorhabditis elegans Proteins genetics, Caenorhabditis elegans Proteins metabolism
- Abstract
Human NIMA-related kinases have primarily been studied for their roles in cell cycle progression (NEK1/2/6/7/9), checkpoint-DNA-damage control (NEK1/2/4/5/10/11), and ciliogenesis (NEK1/4/8). We previously showed that Caenorhabditis elegans NEKL-2 (NEK8/9 homolog) and NEKL-3 (NEK6/7 homolog) regulate apical clathrin-mediated endocytosis (CME) in the worm epidermis and are essential for molting. Here we show that NEKL-2 and NEKL-3 also have distinct roles in controlling endosome function and morphology. Specifically, loss of NEKL-2 led to enlarged early endosomes with long tubular extensions but showed minimal effects on other compartments. In contrast, NEKL-3 depletion caused pronounced defects in early, late, and recycling endosomes. Consistently, NEKL-2 was strongly localized to early endosomes, whereas NEKL-3 was localized to multiple endosomal compartments. Loss of NEKLs also led to variable defects in the recycling of two resident cargoes of the trans-Golgi network (TGN), MIG-14/Wntless and TGN-38/TGN38, which were missorted to lysosomes after NEKL depletion. In addition, defects were observed in the uptake of clathrin-dependent (SMA-6/Type I BMP receptor) and independent cargoes (DAF-4/Type II BMP receptor) from the basolateral surface of epidermal cells after NEKL-2 or NEKL-3 depletion. Complementary studies in human cell lines further showed that siRNA knockdown of the NEKL-3 orthologs NEK6 and NEK7 led to missorting of the mannose 6-phosphate receptor from endosomes. Moreover, in multiple human cell types, depletion of NEK6 or NEK7 disrupted both early and recycling endosomal compartments, including the presence of excess tubulation within recycling endosomes, a defect also observed after NEKL-3 depletion in worms. Thus, NIMA family kinases carry out multiple functions during endocytosis in both worms and humans, consistent with our previous observation that human NEKL-3 orthologs can rescue molting and trafficking defects in C. elegans nekl-3 mutants. Our findings suggest that trafficking defects could underlie some of the proposed roles for NEK kinases in human disease., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2023 Joseph et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
29. The p.Pro2232Leu variant in the ChEL domain of thyroglobulin gene causes intracellular transport disorder and congenital hypothyroidism.
- Author
-
Siffo S, Gomes Pio M, Martínez EB, Lachlan K, Walker J, Weill J, González-Sarmiento R, Rivolta CM, and Targovnik HM
- Subjects
- Animals, Humans, Rats, HEK293 Cells, Thyroglobulin genetics, Thyroglobulin metabolism, Protein Transport genetics, Congenital Hypothyroidism genetics, Goiter
- Abstract
Thyroglobulin (TG), the predominant glycoprotein of the thyroid gland, functions as matrix protein in thyroid hormonegenesis. TG deficiency results in thyroid dyshormonogenesis. These variants produce a heterogeneous spectrum of congenital goitre, with an autosomal recessive mode of inheritance. The purpose of this study was to identify and functionally characterize new variants in the TG gene in order to increase the understanding of the molecular mechanisms responsible for thyroid dyshormonogenesis. A total of four patients from two non-consanguineous families with marked alteration of TG synthesis were studied. The two families were previously analysed in our laboratory, only one deleterious allele, in each one, was detected after sequencing the TG gene (c.2359 C > T [p.Arg787*], c.5560 G > T [p.Glu1854*]). These findings were confirmed in the present studies by Next-Generation Sequencing. The single nucleotide coding variants of the TG gene were then analyzed to predict the possible variant causing the disease. The p.Pro2232Leu (c.6695 C > T), identified in both families, showing a low frequency population in gnomAD v2.1.1 database and protein homology, amino acid prediction, and 3D modeling analysis predict a potential pathogenic effect of this variant. We also transiently express p.Pro2232Leu in a full-length rat TG cDNA clone and confirmed that this point variant was sufficient to cause intracellular retention of mutant TG in HEK293T cells. Consequently, each family carried a compound heterozygous for p.Arg787*/p.Pro2232Leu or p.Glu1854*/p.Pro2232Leu variants. In conclusion, our results confirm the pathophysiological importance of altered TG folding as a consequence of missense variants located in the ChEL domain of TG., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
30. Newcastle Disease Virus Manipulates Mitochondrial MTHFD2-Mediated Nucleotide Metabolism for Virus Replication.
- Author
-
Tang N, Chen P, Zhao C, Liu P, Tan L, Song C, Qiu X, Liao Y, Liu X, Luo T, Sun Y, and Ding C
- Subjects
- Animals, Nucleotides metabolism, Serine metabolism, Cell Line, A549 Cells, Humans, Mesocricetus, Gene Knockdown Techniques, Protein Transport genetics, Mitochondria enzymology, Up-Regulation physiology, Methylenetetrahydrofolate Dehydrogenase (NADP) genetics, Methylenetetrahydrofolate Dehydrogenase (NADP) metabolism, Newcastle Disease enzymology, Newcastle Disease physiopathology, Newcastle Disease virology, Newcastle disease virus genetics, Newcastle disease virus metabolism, Virus Replication genetics
- Abstract
Viruses require host cell metabolic reprogramming to satisfy their replication demands; however, the mechanism by which the Newcastle disease virus (NDV) remodels nucleotide metabolism to support self-replication remains unknown. In this study, we demonstrate that NDV relies on the oxidative pentose phosphate pathway (oxPPP) and the folate-mediated one-carbon metabolic pathway to support replication. In concert with [1,2-
13 C2 ] glucose metabolic flow, NDV used oxPPP to promote pentose phosphate synthesis and to increase antioxidant NADPH production. Metabolic flux experiments using [2,3,3-2 H] serine revealed that NDV increased one-carbon (1C) unit synthesis flux through the mitochondrial 1C pathway. Interestingly, methylenetetrahydrofolate dehydrogenase (MTHFD2) was upregulated as a compensatory mechanism for insufficient serine availability. Unexpectedly, direct knockdown of enzymes in the one-carbon metabolic pathway, except for cytosolic MTHFD1, significantly inhibited NDV replication. Specific complementation rescue experiments on small interfering RNA (siRNA)-mediated knockdown further revealed that only a knockdown of MTHFD2 strongly restrained NDV replication and was rescued by formate and extracellular nucleotides. These findings indicated that NDV replication relies on MTHFD2 to maintain nucleotide availability. Notably, nuclear MTHFD2 expression was increased during NDV infection and could represent a pathway by which NDV steals nucleotides from the nucleus. Collectively, these data reveal that NDV replication is regulated by the c-Myc-mediated 1C metabolic pathway and that the mechanism of nucleotide synthesis for viral replication is regulated by MTHFD2. IMPORTANCE Newcastle disease virus (NDV) is a dominant vector for vaccine and gene therapy that accommodates foreign genes well but can only infect mammalian cells that have undergone cancerous transformation. Understanding the remodeling of nucleotide metabolic pathways in host cells by NDV proliferation provides a new perspective for the precise use of NDV as a vector or in antiviral research. In this study, we demonstrated that NDV replication is strictly dependent on pathways involved in redox homeostasis in the nucleotide synthesis pathway, including the oxPPP and the mitochondrial one-carbon pathway. Further investigation revealed the potential involvement of NDV replication-dependent nucleotide availability in promoting MTHFD2 nuclear localization. Our findings highlight the differential dependence of NDV on enzymes for one-carbon metabolism, and the unique mechanism of action of MTHFD2 in viral replication, thereby providing a novel target for antiviral or oncolytic virus therapy.- Published
- 2023
- Full Text
- View/download PDF
31. Unraveling the intricate cargo-BBSome coupling mechanism at the ciliary tip.
- Author
-
Liu YX, Li WJ, Zhang RK, Sun SN, and Fan ZC
- Subjects
- Humans, Protein Transport genetics, Hedgehog Proteins metabolism, Membrane Proteins metabolism, Guanosine Triphosphate metabolism, Flagella metabolism, Cilia metabolism, Bardet-Biedl Syndrome genetics
- Abstract
Certain ciliary transmembrane and membrane-tethered signaling proteins migrate from the ciliary tip to base via retrograde intraflagellar transport (IFT), essential for maintaining their ciliary dynamics to enable cells to sense and transduce extracellular stimuli inside the cell. During this process, the BBSome functions as an adaptor between retrograde IFT trains and these signaling protein cargoes. The Arf-like 13 (ARL13) small GTPase resembles ARL6/BBS3 in facilitating these signaling cargoes to couple with the BBSome at the ciliary tip prior to loading onto retrograde IFT trains for transporting towards the ciliary base, while the molecular basis for how this intricate coupling event happens remains elusive. Here, we report that Chlamydomonas ARL13 only in a GTP-bound form (ARL13
GTP ) anchors to the membrane for diffusing into cilia. Upon entering cilia, ARL13 undergoes GTPase cycle for shuttling between the ciliary membrane (ARL13GTP ) and matrix (ARL13GDP ). To achieve this goal, the ciliary membrane-anchored BBS3GTP binds the ciliary matrix-residing ARL13GDP to activate the latter as an ARL13 guanine nucleotide exchange factor. At the ciliary tip, ARL13GTP recruits the ciliary matrix-residing and post-remodeled BBSome as an ARL13 effector to anchor to the ciliary membrane. This makes the BBSome spatiotemporally become available for the ciliary membrane-tethered phospholipase D (PLD) to couple with. Afterward, ARL13GTP hydrolyzes GTP for releasing the PLD-laden BBSome to load onto retrograde IFT trains. According to this model, hedgehog signaling defects associated with ARL13b and BBS3 mutations in humans could be satisfactorily explained, providing us a mechanistic understanding behind BBSome-cargo coupling required for proper ciliary signaling.- Published
- 2023
- Full Text
- View/download PDF
32. Influence of glycoprotein MUC1 on trafficking of the Ca 2+ -selective ion channels, TRPV5 and TRPV6, and on in vivo calcium homeostasis.
- Author
-
Al-Bataineh MM, Kinlough CL, Marciszyn A, Lam T, Ye L, Kidd K, Maggiore JC, Poland PA, Kmoch S, Bleyer A, Bain DJ, Montalbetti N, Kleyman TR, Hughey RP, and Ray EC
- Subjects
- Animals, Female, Humans, Mice, Cell Membrane metabolism, Cells, Cultured, Epithelial Cells metabolism, Sex Factors, Mutation, Protein Transport genetics, Calcium blood, Calcium metabolism, Calcium urine, Mucin-1 genetics, Mucin-1 metabolism, TRPV Cation Channels metabolism
- Abstract
Polymorphism of the gene encoding mucin 1 (MUC1) is associated with skeletal and dental phenotypes in human genomic studies. Animals lacking MUC1 exhibit mild reduction in bone density. These phenotypes could be a consequence of modulation of bodily Ca homeostasis by MUC1, as suggested by the previous observation that MUC1 enhances cell surface expression of the Ca
2+ -selective channel, TRPV5, in cultured unpolarized cells. Using biotinylation of cell surface proteins, we asked whether MUC1 influences endocytosis of TRPV5 and another Ca2+ -selective TRP channel, TRPV6, in cultured polarized epithelial cells. Our results indicate that MUC1 reduces endocytosis of both channels, enhancing cell surface expression. Further, we found that mice lacking MUC1 lose apical localization of TRPV5 and TRPV6 in the renal tubular and duodenal epithelium. Females, but not males, lacking MUC1 exhibit reduced blood Ca2+ . However, mice lacking MUC1 exhibited no differences in basal urinary Ca excretion or Ca retention in response to PTH receptor signaling, suggesting compensation by transport mechanisms independent of TRPV5 and TRPV6. Finally, humans with autosomal dominant tubulointerstitial kidney disease due to frame-shift mutation of MUC1 (ADTKD-MUC1) exhibit reduced plasma Ca concentrations compared to control individuals with mutations in the gene encoding uromodulin (ADTKD-UMOD), consistent with MUC1 haploinsufficiency causing reduced bodily Ca2+ . In summary, our results provide further insight into the role of MUC1 in Ca2+ -selective TRP channel endocytosis and the overall effects on Ca concentrations., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
33. VPS35 promotes cell proliferation via EGFR recycling and enhances EGFR inhibitors response in gastric cancer.
- Author
-
Yu J, Feng H, Sang Q, Li F, Chen M, Yu B, Xu Z, Pan T, Wu X, Hou J, Zhu Z, Yan C, Su L, Li J, and Liu B
- Subjects
- Humans, Carrier Proteins metabolism, Cell Proliferation, ErbB Receptors antagonists & inhibitors, ErbB Receptors genetics, ErbB Receptors metabolism, Protein Transport drug effects, Protein Transport genetics, Stomach Neoplasms genetics, Vesicular Transport Proteins genetics, Vesicular Transport Proteins metabolism
- Abstract
Background: Vacuolar protein sorting-associated protein 35 (VPS35) is a core component of the retromer complex which mediates intracellular protein transport. It is well known that dysfunctional VPS35 functions in the accumulation of pathogenic proteins. In our previous study, VPS35 was found to be a potential gene related to poor prognosis in gastric cancer. However, the biological functions of VPS35 in gastric cancer remain unclear., Methods: Cell viability assays were performed to examine whether VPS35 affected cell proliferation. Immunoprecipitation and biotin assays showed that VPS35 bound to epidermal growth factor receptor (EGFR) in the cytoplasm and recycled it to the cell surface. Patient-derived xenografts and organoids were used to evaluate the effect of VPS35 on the response of gastric cancer to EGFR inhibitors., Findings: VPS35 expression levels were upregulated in tumour tissues and correlated with local tumour invasion and poor survival in patients with gastric cancer. VPS35 promoted cell proliferation and increased tumour growth. Mechanistically, VPS35 selectively bound to endocytosed EGFR in early endosomes and recycled it back to the cell surface, leading to the downstream activation of the ERK1/2 pathway. We also found that high VPS35 expression levels increased the sensitivity of the xenograft and organoid models to EGFR inhibitors., Interpretation: VPS35 promotes cell proliferation by recycling EGFR to the cell surface, amplifying the network of receptor trafficking. VPS35 expression levels are positively correlated with gastric cancer sensitivity to EGFR inhibitors, which offers a potential method to stratify patients for EGFR inhibitor utilisation., Funding: National Natural Science Foundation of China., Competing Interests: Declaration of interests All the authors have declared no conflicts of interests., (Copyright © 2023 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
34. Decreasing mutant ATXN1 nuclear localization improves a spectrum of SCA1-like phenotypes and brain region transcriptomic profiles.
- Author
-
Handler HP, Duvick L, Mitchell JS, Cvetanovic M, Reighard M, Soles A, Mather KB, Rainwater O, Serres S, Nichols-Meade T, Coffin SL, You Y, Ruis BL, O'Callaghan B, Henzler C, Zoghbi HY, and Orr HT
- Subjects
- Animals, Mice, Brain metabolism, Cerebellum metabolism, Disease Models, Animal, Mice, Transgenic, Nerve Tissue Proteins genetics, Phenotype, Protein Transport genetics, Purkinje Cells metabolism, Ataxin-1 genetics, Ataxin-1 metabolism, Spinocerebellar Ataxias genetics, Spinocerebellar Ataxias metabolism, Transcriptome
- Abstract
Spinocerebellar ataxia type 1 (SCA1) is a dominant trinucleotide repeat neurodegenerative disease characterized by motor dysfunction, cognitive impairment, and premature death. Degeneration of cerebellar Purkinje cells is a frequent and prominent pathological feature of SCA1. We previously showed that transport of ATXN1 to Purkinje cell nuclei is required for pathology, where mutant ATXN1 alters transcription. To examine the role of ATXN1 nuclear localization broadly in SCA1-like disease pathogenesis, CRISPR-Cas9 was used to develop a mouse with an amino acid alteration (K772T) in the nuclear localization sequence of the expanded ATXN1 protein. Characterization of these mice indicates that proper nuclear localization of mutant ATXN1 contributes to many disease-like phenotypes including motor dysfunction, cognitive deficits, and premature lethality. RNA sequencing analysis of genes with expression corrected to WT levels in Atxn1
175QK772T/2Q mice indicates that transcriptomic aspects of SCA1 pathogenesis differ between the cerebellum, brainstem, cerebral cortex, hippocampus, and striatum., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
35. Surf4, cargo trafficking, lipid metabolism, and therapeutic implications.
- Author
-
Shen Y, Gu HM, Qin S, and Zhang DW
- Subjects
- Humans, Golgi Apparatus metabolism, Proprotein Convertase 9 metabolism, Protein Transport genetics, Protein Transport physiology, Lipid Metabolism, Membrane Proteins metabolism
- Abstract
Surfeit 4 is a polytopic transmembrane protein that primarily resides in the endoplasmic reticulum (ER) membrane. It is ubiquitously expressed and functions as a cargo receptor, mediating cargo transport from the ER to the Golgi apparatus via the canonical coat protein complex II (COPII)-coated vesicles or specific vesicles. It also participates in ER-Golgi protein trafficking through a tubular network. Meanwhile, it facilitates retrograde transportation of cargos from the Golgi apparatus to the ER through COPI-coated vesicles. Surf4 can selectively mediate export of diverse cargos, such as PCSK9 very low-density lipoprotein (VLDL), progranulin, α1-antitrypsin, STING, proinsulin, and erythropoietin. It has been implicated in facilitating VLDL secretion, promoting cell proliferation and migration, and increasing replication of positive-strand RNA viruses. Therefore, Surf4 plays a crucial role in various physiological and pathophysiological processes and emerges as a promising therapeutic target. However, the molecular mechanisms by which Surf4 selectively sorts diverse cargos for ER-Golgi protein trafficking remain elusive. Here, we summarize the most recent advances in Surf4, focusing on its role in lipid metabolism., (© The Author(s) (2022). Published by Oxford University Press on behalf of Journal of Molecular Cell Biology, CEMCS, CAS.)
- Published
- 2023
- Full Text
- View/download PDF
36. A deleterious Sar1c variant in rice inhibits export of seed storage proteins from the endoplasmic reticulum.
- Author
-
Bao X, Wang Y, Qi Y, Lei C, Wang Y, Pan T, Yu M, Zhang Y, Wu H, Zhang P, Ji Y, Yang H, Jiang X, Jing R, Yan M, Zhang B, Gu C, Zhu J, Hao Y, Lei J, Zhang S, Chen X, Chen R, Sun Y, Zhu Y, Zhang X, Jiang L, Visser RGF, Ren Y, Wang Y, and Wan J
- Subjects
- Protein Transport genetics, Glutens genetics, Endoplasmic Reticulum metabolism, Seed Storage Proteins metabolism, Oryza genetics
- Abstract
Key Message: We identified a dosage-dependent dominant negative form of Sar1c, which confirms the essential role of COPII system in mediating ER export of storage proteins in rice endosperm. Higher plants accumlate large amounts of seed storage proteins (SSPs). However, mechanisms underlying SSP trafficking are largely unknown, especially the ER-Golgi anterograde process. Here, we showed that a rice glutelin precursor accumulation13 (gpa13) mutant exhibited floury endosperm and overaccumulated glutelin precursors, which phenocopied the reported RNAi-Sar1abc line. Molecular cloning revealed that the gpa13 allele encodes a mutated Sar1c (mSar1c) with a deletion of two conserved amino acids Pro134 and Try135. Knockdown or knockout of Sar1c alone caused no obvious phenotype, while overexpression of mSar1c resulted in seedling lethality similar to the gpa13 mutant. Transient expression experiment in tobacco combined with subcellular fractionation experiment in gpa13 demonstrated that the expression of mSar1c affects the subcellular distribution of all Sar1 isoforms and Sec23c. In addition, mSar1c failed to interact with COPII component Sec23. Conversely, mSar1c competed with Sar1a/b/d to interact with guanine nucleotide exchange factor Sec12. Together, we identified a dosage-dependent dominant negative form of Sar1c, which confirms the essential role of COPII system in mediating ER export of storage proteins in rice endosperm., (© 2022. The Author(s), under exclusive licence to Springer Nature B.V.)
- Published
- 2023
- Full Text
- View/download PDF
37. Regulation of transferrin receptor trafficking by optineurin and its disease-associated mutants.
- Author
-
Moharir SC, Sirohi K, and Swarup G
- Subjects
- Humans, Biological Transport, Endosomes metabolism, Iron metabolism, Protein Transport genetics, Transferrins metabolism, Cell Cycle Proteins genetics, Endocytosis genetics, Membrane Transport Proteins genetics, Receptors, Transferrin metabolism
- Abstract
Transferrin receptor (TFRC) is a transmembrane protein that plays a crucial role in mediating homeostasis of iron in the cell. The binding of transferrin (that is bound to iron) to TFRC at the cell membrane generally starts endocytosis of TFRC-transferrin complex, which leads to formation of vesicles that are positive for TFRC. These vesicles travel to the early endosomes and later to the endocytic recycling compartment. Release of iron occurs in the early endosomes because of acidic pH. Major fraction of the transferrin and TFRC is transported back to the cell membrane; however, a minor fraction of it is transported to lysosomes through the process of autophagy. Optineurin (OPTN) is a multi-functional adaptor protein that plays a pivotal role in the control of TFRC trafficking, recycling and autophagy dependent degradation. Optineurin also plays a role in cargo-selective and non-selective autophagy. Here, we review our understanding of the function of OPTN in regulating TFRC trafficking, recycling and autophagy dependent degradation. We also discuss the mechanisms by which certain disease-associated mutations of OPTN alter these processes., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
38. The PTEX Pore Component EXP2 Is Important for Intrahepatic Development during the Plasmodium Liver Stage.
- Author
-
Hussain T, Linera-Gonzalez J, Beck JM, Fierro MA, Mair GR, Smith RC, and Beck JR
- Subjects
- Carcinoma, Hepatocellular, Liver Neoplasms, Plasmodium falciparum genetics, Protein Transport genetics, Protein Transport physiology, RNA, Catalytic metabolism, Animals, Mice, Erythrocytes metabolism, Erythrocytes parasitology, Malaria genetics, Malaria metabolism, Malaria parasitology, Plasmodium berghei genetics, Plasmodium berghei metabolism, Protozoan Proteins genetics, Protozoan Proteins metabolism, Hepatocytes metabolism, Hepatocytes parasitology
- Abstract
During vertebrate infection, obligate intracellular malaria parasites develop within a parasitophorous vacuole, which constitutes the interface between the parasite and its hepatocyte or erythrocyte host cells. To traverse this barrier, Plasmodium spp. utilize a dual-function pore formed by EXP2 for nutrient transport and, in the context of the PTEX translocon, effector protein export across the vacuole membrane. While critical to blood-stage survival, less is known about EXP2/PTEX function in the liver stage, although major differences in the export mechanism are suggested by absence of the PTEX unfoldase HSP101 in the intrahepatic vacuole. Here, we employed the glucosamine-activated glmS ribozyme to study the role of EXP2 during Plasmodium berghei liver-stage development in hepatoma cells. Insertion of the glmS sequence into the exp2 3' untranslated region (UTR) enabled glucosamine-dependent depletion of EXP2 after hepatocyte invasion, allowing separation of EXP2 function during intrahepatic development from a recently reported role in hepatocyte invasion. Postinvasion EXP2 knockdown reduced parasite size and largely abolished expression of the mid- to late-liver-stage marker LISP2. As an orthogonal approach to monitor development, EXP2- glmS parasites and controls were engineered to express nanoluciferase. Activation of glmS after invasion substantially decreased luminescence in hepatoma monolayers and in culture supernatants at later time points corresponding to merosome detachment, which marks the culmination of liver-stage development. Collectively, our findings extend the utility of the glmS ribozyme to study protein function in the liver stage and reveal that EXP2 is important for intrahepatic parasite development, indicating that PTEX components also function at the hepatocyte-parasite interface. IMPORTANCE After the mosquito bite that initiates a Plasmodium infection, parasites first travel to the liver and develop in hepatocytes. This liver stage is asymptomatic but necessary for the parasite to transition to the merozoite form, which infects red blood cells and causes malaria. To take over their host cells, avoid immune defenses, and fuel their growth, these obligately intracellular parasites must import nutrients and export effector proteins across a vacuole membrane in which they reside. In the blood stage, these processes depend on a translocon called PTEX, but it is unclear if PTEX also functions during the liver stage. Here, we adapted the glmS ribozyme to control expression of EXP2, the membrane pore component of PTEX, during the liver stage of the rodent malaria parasite Plasmodium berghei. Our results show that EXP2 is important for intracellular development in the hepatocyte, revealing that PTEX components are also functionally important during liver-stage infection.
- Published
- 2022
- Full Text
- View/download PDF
39. Enhanced myogenesis through lncFAM-mediated recruitment of HNRNPL to the MYBPC2 promoter.
- Author
-
Chang MW, Yang JH, Tsitsipatis D, Yang X, Martindale JL, Munk R, Pandey PR, Banskota N, Romero B, Batish M, Piao Y, Mazan-Mamczarz K, De S, Abdelmohsen K, Wilson GM, and Gorospe M
- Subjects
- Humans, Muscle Fibers, Skeletal metabolism, Myoblasts cytology, Myoblasts metabolism, Gene Silencing, Protein Transport genetics, Heterogeneous-Nuclear Ribonucleoprotein L genetics, Heterogeneous-Nuclear Ribonucleoprotein L metabolism, Muscle Development genetics, RNA, Long Noncoding genetics, RNA, Long Noncoding metabolism, Transcription, Genetic genetics, Promoter Regions, Genetic
- Abstract
The mammalian transcriptome comprises a vast family of long noncoding (lnc)RNAs implicated in physiologic processes such as myogenesis, through which muscle forms during embryonic development and regenerates in the adult. However, the specific molecular mechanisms by which lncRNAs regulate human myogenesis are poorly understood. Here, we identified a novel muscle-specific lncRNA, lncFAM71E1-2:2 (lncFAM), which increased robustly during early human myogenesis. Overexpression of lncFAM promoted differentiation of human myoblasts into myotubes, while silencing lncFAM suppressed this process. As lncFAM resides in the nucleus, chromatin isolation by RNA purification followed by mass spectrometry (ChIRP-MS) analysis was employed to identify the molecular mechanisms whereby it might promote myogenesis. Analysis of lncFAM-interacting proteins revealed that lncFAM recruited the RNA-binding protein HNRNPL to the promoter of MYBPC2, in turn increasing MYBPC2 mRNA transcription and enhancing production of the myogenic protein MYBPC2. These results highlight a mechanism whereby a novel ribonucleoprotein complex, lncFAM-HNRNPL, elevates MYBPC2 expression transcriptionally to promote myogenesis., (Published by Oxford University Press on behalf of Nucleic Acids Research 2022.)
- Published
- 2022
- Full Text
- View/download PDF
40. Pushing and pulling proteins into the yeast secretory pathway enhances recombinant protein secretion.
- Author
-
Zahrl RJ, Prielhofer R, Ata Ö, Baumann K, Mattanovich D, and Gasser B
- Subjects
- Recombinant Proteins, Protein Transport genetics, Metabolic Engineering, Pichia genetics, Pichia metabolism, Secretory Pathway genetics
- Abstract
Yeasts and especially Pichia pastoris (syn Komagataella spp.) are popular microbial expression systems for the production of recombinant proteins. One of the key advantages of yeast host systems is their ability to secrete the recombinant protein into the culture media. However, secretion of some recombinant proteins is less efficient. These proteins include antibody fragments such as Fabs or scFvs. We have recently identified translocation of nascent Fab fragments from the cytosol into the endoplasmic reticulum (ER) as one major bottleneck. Conceptually, this bottleneck requires engineering to increase the flux of recombinant proteins at the translocation step by pushing on the cytosolic side and pulling on the ER side. This engineering strategy is well-known in the field of metabolic engineering. To apply the push-and-pull strategy to recombinant protein secretion, we chose to modulate the cytosolic and ER Hsp70 cycles, which have a key impact on the translocation process. After identifying the relevant candidate factors of the Hsp70 cycles, we combined the push-and-pull factors in a single strain and achieved synergistic effects for antibody fragment secretion. With this concept we were able to successfully engineer strains and improve protein secretion up to 5-fold for different model protein classes. Overall, titers of more than 1.3 g/L Fab and scFv were reached in bioreactor cultivations., Competing Interests: Declaration of competing interest None., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
41. CEP19-RABL2-IFT-B axis controls BBSome-mediated ciliary GPCR export.
- Author
-
Zhou Z, Katoh Y, and Nakayama K
- Subjects
- Cell Cycle Proteins metabolism, Cytoskeletal Proteins metabolism, Flagella metabolism, GTP Phosphohydrolases metabolism, Guanosine Triphosphate metabolism, Humans, Membrane Proteins metabolism, Protein Transport genetics, Bardet-Biedl Syndrome genetics, Bardet-Biedl Syndrome metabolism, Cilia metabolism
- Abstract
The intraflagellar transport (IFT) machinery mediates the import and export of ciliary proteins across the ciliary gate, as well as bidirectional protein trafficking within cilia. In addition to ciliary anterograde protein trafficking, the IFT-B complex participates in the export of membrane proteins together with the BBSome, which consists of eight subunits encoded by the causative genes of Bardet-Biedl syndrome (BBS). The IFT25-IFT27/BBS19 dimer in the IFT-B complex constitutes its interface with the BBSome. We show here that IFT25-IFT27 and the RABL2 GTPase bind the IFT74/BBS22-IFT81 dimer of the IFT-B complex in a mutually exclusive manner. Cells expressing GTP-locked RABL2 [RABL2(Q80L)], but not wild-type RABL2, phenocopied IFT27 -knockout cells, that is, they demonstrated BBS-associated ciliary defects, including accumulation of LZTFL1/BBS17 and the BBSome within cilia and the suppression of export of the ciliary GPCRs GPR161 and Smoothened. RABL2(Q80L) enters cilia in a manner dependent on the basal body protein CEP19, but its entry into cilia is not necessary for causing BBS-associated ciliary defects. These observations suggest that GTP-bound RABL2 is likely to be required for recruitment of the IFT-B complex to the ciliary base, where it is replaced with IFT25-IFT27.
- Published
- 2022
- Full Text
- View/download PDF
42. RABENOSYN separation-of-function mutations uncouple endosomal recycling from lysosomal degradation, causing a distinct Mendelian disorder.
- Author
-
Paul F, Ng C, Mohamad Sahari UB, Nafissi S, Nilipoor Y, Tavasoli AR, Bonnard C, Wong PM, Nabavizadeh N, Altunoğlu U, Estiar MA, Majoie CB, Lee H, Nelson SF, Gan-Or Z, Rouleau GA, Van Veldhoven PP, Massie R, Hennekam RC, Kariminejad A, and Reversade B
- Subjects
- Humans, Alleles, Lysosomes genetics, Lysosomes metabolism, Mutation, Protein Transport genetics, Endosomes genetics, Endosomes metabolism, Intellectual Disability genetics, Vesicular Transport Proteins genetics
- Abstract
Rabenosyn (RBSN) is a conserved endosomal protein necessary for regulating internalized cargo. Here, we present clinical, genetic, cellular and biochemical evidence that two distinct RBSN missense variants are responsible for a novel Mendelian disorder consisting of progressive muscle weakness, facial dysmorphisms, ophthalmoplegia and intellectual disability. Using exome sequencing, we identified recessively acting germline alleles p.Arg180Gly and p.Gly183Arg, which are both situated in the FYVE domain of RBSN. We find that these variants abrogate binding to its cognate substrate phosphatidylinositol 3-phosphate (PI3P) and thus prevent its translocation to early endosomes. Although the endosomal recycling pathway was unaltered, mutant p.Gly183Arg patient fibroblasts show accumulation of cargo tagged for lysosomal degradation. Our results suggest that these variants are separation-of-function alleles, which cause a delay in endosomal maturation without affecting cargo recycling. We conclude that distinct germline mutations in RBSN cause non-overlapping phenotypes with specific and discrete endolysosomal cellular defects., (© The Author(s) 2022. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
43. Decreased Expression of the Slc31a1 Gene and Cytoplasmic Relocalization of Membrane CTR1 Protein in Renal Epithelial Cells: A Potent Protective Mechanism against Copper Nephrotoxicity in a Mouse Model of Menkes Disease.
- Author
-
Haberkiewicz O, Lipiński P, Starzyński RR, Jończy A, Kurowska P, Ogórek M, Bednarz A, Herman S, Hatala D, Grzmil P, Rajfur Z, Baster Z, and Lenartowicz M
- Subjects
- Animals, Cation Transport Proteins genetics, Cation Transport Proteins metabolism, Cell Membrane genetics, Cell Membrane metabolism, Copper-Transporting ATPases genetics, Copper-Transporting ATPases metabolism, Cytoplasm genetics, Cytoplasm metabolism, Disease Models, Animal, Gene Expression, Membrane Proteins genetics, Membrane Proteins metabolism, Mice, Protein Transport genetics, Protein Transport physiology, RNA, Messenger metabolism, SLC31 Proteins genetics, SLC31 Proteins metabolism, Copper metabolism, Copper toxicity, Copper Transporter 1 genetics, Copper Transporter 1 metabolism, Epithelial Cells metabolism, Kidney Tubules, Proximal metabolism, Menkes Kinky Hair Syndrome etiology, Menkes Kinky Hair Syndrome genetics, Menkes Kinky Hair Syndrome metabolism
- Abstract
Kidneys play an especial role in copper redistribution in the organism. The epithelial cells of proximal tubules perform the functions of both copper uptake from the primary urine and release to the blood. These cells are equipped on their apical and basal membrane with copper transporters CTR1 and ATP7A. Mosaic mutant mice displaying a functional dysfunction of ATP7A are an established model of Menkes disease. These mice exhibit systemic copper deficiency despite renal copper overload, enhanced by copper therapy, which is indispensable for their life span extension. The aim of this study was to analyze the expression of Slc31a1 and Slc31a2 genes (encoding CTR1/CTR2 proteins) and the cellular localization of the CTR1 protein in suckling, young and adult mosaic mutants. Our results indicate that in the kidney of both intact and copper-injected 14-day-old mutants showing high renal copper content, CTR1 mRNA level is not up-regulated compared to wild-type mice given a copper injection. The expression of the Slc31a1 gene in 45-day-old mice is even reduced compared with intact wild-type animals. In suckling and young copper-injected mutants, the CTR1 protein is relocalized from the apical membrane to the cytoplasm of epithelial cells of proximal tubules, the process which prevents copper transport from the primary urine and, thus, protects cells against copper toxicity.
- Published
- 2022
- Full Text
- View/download PDF
44. The intraflagellar transport protein IFT52 associated with short-rib thoracic dysplasia is essential for ciliary function in osteogenic differentiation in vitro and for sensory perception in Drosophila.
- Author
-
Guleria VS, Parit R, Quadri N, Das R, and Upadhyai P
- Subjects
- Animals, Carrier Proteins metabolism, Cilia metabolism, Drosophila metabolism, Humans, Intracellular Signaling Peptides and Proteins, Mice, Perception, Protein Transport genetics, Ribs metabolism, Hedgehog Proteins genetics, Hedgehog Proteins metabolism, Osteogenesis genetics
- Abstract
Primary cilia are non-motile sensory cell-organelle that are essential for organismal development, differentiation, and postnatal homeostasis. Their biogenesis and function are mediated by the intraflagellar transport (IFT) system. Pathogenic variants in IFT52, a central component of the IFT-B complex is associated with short-rib thoracic dysplasia with or without polydactyly 16 (SRTD16), with major skeletal manifestations, in addition to other features. Here we sought to examine the role of IFT52 in osteoblast differentiation. Using lentiviral shRNA interference Ift52 was depleted in C3H10T1/2 mouse mesenchymal stem cells. This led to the disruption of the IFT-B anterograde trafficking machinery that impaired primary ciliogenesis and blocked osteogenic differentiation. In Ift52 silenced cells, Hedgehog (Hh) pathway upregulation during osteogenesis was attenuated and despite Smoothened Agonist (SAG) based Hh activation, osteogenic differentiation was incompletely restored. Further we investigated IFT52 activity in Drosophila, wherein the only ciliated somatic cells are the bipolar sensory neurons of the peripheral nervous system. Knockdown of IFT52 in Drosophila neuronal tissues reduced lifespan with the loss of embryonic chordotonal cilia, and produced severe locomotion, auditory and proprioceptive defects in larva and adults. Together these findings improve our knowledge of the role of IFT52 in various physiological contexts and its associated human disorder., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
45. Deletion of CEP164 in mouse photoreceptors post-ciliogenesis interrupts ciliary intraflagellar transport (IFT).
- Author
-
Reed M, Takemaru KI, Ying G, Frederick JM, and Baehr W
- Subjects
- Animals, Cilia metabolism, Mice, Protein Transport genetics, Retinal Cone Photoreceptor Cells, Tamoxifen, Basal Bodies metabolism, Fluorescent Dyes metabolism
- Abstract
Centrosomal protein of 164 kDa (CEP164) is located at distal appendages of primary cilia and is necessary for basal body (BB) docking to the apical membrane. To investigate the function of photoreceptor CEP164 before and after BB docking, we deleted CEP164 during retina embryonic development (Six3Cre), in postnatal rod photoreceptors (iCre75) and in mature retina using tamoxifen induction (Prom1-ETCre). BBs dock to the cell cortex during postnatal day 6 (P6) to extend a connecting cilium (CC) and an axoneme. P6 retina-specific knockouts (retCep164-/-) are unable to dock BBs, thereby preventing formation of CC or outer segments (OSs). In rod-specific knockouts (rodCep164-/-), Cre expression starts after P7 and CC/OS form. P16 rodCep164-/- rods have nearly normal OS lengths, and maintain OS attachment through P21 despite loss of CEP164. Intraflagellar transport components (IFT88, IFT57 and IFT140) were reduced at P16 rodCep164-/- BBs and CC tips and nearly absent at P21, indicating impaired intraflagellar transport. Nascent OS discs, labeled with a fluorescent dye on P14 and P18 and harvested on P19, showed continued rodCep164-/- disc morphogenesis but absence of P14 discs mid-distally, indicating OS instability. Tamoxifen induction with PROM1ETCre;Cep164F/F (tamCep164-/-) adult mice affected maintenance of both rod and cone OSs. The results suggest that CEP164 is key towards recruitment and stabilization of IFT-B particles at the BB/CC. IFT impairment may be the main driver of ciliary malfunction observed with hypomorphic CEP164 mutations., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2022
- Full Text
- View/download PDF
46. Structural basis of lipoprotein recognition by the bacterial Lol trafficking chaperone LolA.
- Author
-
Kaplan E, Greene NP, Jepson AE, and Koronakis V
- Subjects
- Bacterial Outer Membrane Proteins genetics, Bacterial Outer Membrane Proteins metabolism, Carrier Proteins metabolism, Escherichia coli genetics, Escherichia coli metabolism, Ligands, Lipoproteins metabolism, Models, Molecular, Molecular Chaperones genetics, Molecular Chaperones metabolism, Periplasm metabolism, Protein Structure, Tertiary, Escherichia coli Proteins chemistry, Escherichia coli Proteins genetics, Escherichia coli Proteins metabolism, Periplasmic Binding Proteins chemistry, Periplasmic Binding Proteins genetics, Periplasmic Binding Proteins metabolism, Protein Transport genetics
- Abstract
In gram-negative bacteria, lipoproteins are vital structural components of the outer membrane (OM) and crucial elements of machineries central to the physiology of the cell envelope. A dedicated apparatus, the Lol system, is required for the correct localization of OM lipoproteins and is essential for viability. The periplasmic chaperone LolA is central to this trafficking pathway, accepting triacylated lipoproteins from the inner membrane transporter LolCDE, before carrying them across the periplasm to the OM receptor LolB. Here, we report a crystal structure of liganded LolA, generated in vivo, revealing the molecular details of lipoprotein association. The structure highlights how LolA, initially primed to receive lipoprotein by interaction with LolC, further opens to accommodate the three ligand acyl chains in a precise conformation within its cavity. LolA forms extensive interactions with the acyl chains but not with any residue of the cargo, explaining the chaperone's ability to transport structurally diverse lipoproteins. Structural characterization of a ligandedLolA variant incapable of lipoprotein release reveals aberrant association, demonstrating the importance of the LolCDE-coordinated, sequential opening of LolA for inserting lipoprotein in a manner productive for subsequent trafficking. Comparison with existing structures of LolA in complex with LolC or LolCDE reveals substantial overlap of the lipoprotein and LolC binding sites within the LolA cavity, demonstrating that insertion of lipoprotein acyl chains physically disengages the chaperone protein from the transporter by perturbing interaction with LolC. Taken together, our data provide a key step toward a complete understanding of a fundamentally important trafficking pathway.
- Published
- 2022
- Full Text
- View/download PDF
47. Twin-arginine translocase component TatB performs folding quality control via a chaperone-like activity.
- Author
-
Taw MN, Boock JT, Sotomayor B, Kim D, Rocco MA, Waraho-Zhmayev D, and DeLisa MP
- Subjects
- Arginine metabolism, Escherichia coli genetics, Escherichia coli metabolism, Humans, Molecular Chaperones genetics, Molecular Chaperones metabolism, Protein Sorting Signals, Escherichia coli Proteins chemistry, Escherichia coli Proteins metabolism, Membrane Transport Proteins chemistry, Membrane Transport Proteins metabolism, Protein Folding, Protein Transport genetics, Protein Transport physiology
- Abstract
The twin-arginine translocation (Tat) pathway involves an inbuilt quality control (QC) system that synchronizes the proofreading of substrate protein folding with lipid bilayer transport. However, the molecular details of this QC mechanism remain poorly understood. Here, we hypothesized that the conformational state of Tat substrates is directly sensed by the TatB component of the bacterial Tat translocase. In support of this hypothesis, several TatB variants were observed to form functional translocases in vivo that had compromised QC activity as evidenced by the uncharacteristic export of several misfolded protein substrates. These variants each possessed cytoplasmic membrane-extrinsic domains that were either truncated or mutated in the vicinity of a conserved, highly flexible α-helical domain. In vitro folding experiments revealed that the TatB membrane-extrinsic domain behaved like a general molecular chaperone, transiently binding to highly structured, partially unfolded intermediates of a model protein, citrate synthase, in a manner that prevented its irreversible aggregation and stabilized the active species. Collectively, these results suggest that the Tat translocase may use chaperone-like client recognition to monitor the conformational status of its substrates., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
48. A massively parallel assay accurately discriminates between functionally normal and abnormal variants in a hotspot domain of KCNH2.
- Author
-
Ng CA, Ullah R, Farr J, Hill AP, Kozek KA, Vanags LR, Mitchell DW, Kroncke BM, and Vandenberg JI
- Subjects
- Alleles, Death, Sudden, Cardiac, Humans, Protein Transport genetics, ERG1 Potassium Channel genetics, ERG1 Potassium Channel metabolism, Ether-A-Go-Go Potassium Channels genetics, Ether-A-Go-Go Potassium Channels metabolism, Long QT Syndrome genetics, Long QT Syndrome metabolism
- Abstract
Many genes, including KCNH2, contain "hotspot" domains associated with a high density of variants associated with disease. This has led to the suggestion that variant location can be used as evidence supporting classification of clinical variants. However, it is not known what proportion of all potential variants in hotspot domains cause loss of function. Here, we have used a massively parallel trafficking assay to characterize all single-nucleotide variants in exon 2 of KCNH2, a known hotspot for variants that cause long QT syndrome type 2 and an increased risk of sudden cardiac death. Forty-two percent of KCNH2 exon 2 variants caused at least 50% reduction in protein trafficking, and 65% of these trafficking-defective variants exerted a dominant-negative effect when co-expressed with a WT KCNH2 allele as assessed using a calibrated patch-clamp electrophysiology assay. The massively parallel trafficking assay was more accurate (AUC of 0.94) than bioinformatic prediction tools (REVEL and CardioBoost, AUC of 0.81) in discriminating between functionally normal and abnormal variants. Interestingly, over half of variants in exon 2 were found to be functionally normal, suggesting a nuanced interpretation of variants in this "hotspot" domain is necessary. Our massively parallel trafficking assay can provide this information prospectively., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 American Society of Human Genetics. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
49. Engineering heterologous enzyme secretion in Yarrowia lipolytica.
- Author
-
Wang W and Blenner MA
- Subjects
- Endoplasmic Reticulum metabolism, Muramidase genetics, Muramidase metabolism, Protein Transport genetics, Secretory Pathway genetics, Yarrowia genetics, Yarrowia metabolism
- Abstract
Background: Eukaryotic cells are often preferred for the production of complex enzymes and biopharmaceuticals due to their ability to form post-translational modifications and inherent quality control system within the endoplasmic reticulum (ER). A non-conventional yeast species, Yarrowia lipolytica, has attracted attention due to its high protein secretion capacity and advanced secretory pathway. Common means of improving protein secretion in Y. lipolytica include codon optimization, increased gene copy number, inducible expression, and secretory tag engineering. In this study, we develop effective strategies to enhance protein secretion using the model heterologous enzyme T4 lysozyme., Results: By engineering the commonly used native lip2prepro secretion signal, we have successfully improved secreted T4 lysozyme titer by 17-fold. Similar improvements were measured for other heterologous proteins, including hrGFP and [Formula: see text]-amylase. In addition to secretion tag engineering, we engineered the secretory pathway by expanding the ER and co-expressing heterologous enzymes in the secretion tag processing pathway, resulting in combined 50-fold improvement in T4 lysozyme secretion., Conclusions: Overall, our combined strategies not only proved effective in improving the protein production in Yarrowia lipolytica, but also hint the possible existence of a different mechanism of secretion regulation in ER and Golgi body in this non-conventional yeast., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
50. Acetylation dependent translocation of EWSR1 regulates CHK2 alternative splicing in response to DNA damage.
- Author
-
Zhang T, Wang Z, Liu M, Liu L, Yang X, Zhang Y, Bie J, Li Y, Ren M, Song C, Wang W, Tan H, and Luo J
- Subjects
- Acetylation, Alternative Splicing genetics, Histone Deacetylases genetics, Histone Deacetylases metabolism, Humans, Oncogene Proteins, Fusion genetics, Protein Transport genetics, Protein Transport physiology, Checkpoint Kinase 2 genetics, Checkpoint Kinase 2 metabolism, DNA Damage genetics, DNA Damage physiology, RNA-Binding Protein EWS genetics, RNA-Binding Protein EWS metabolism, Sarcoma, Ewing genetics
- Abstract
Ewing sarcoma breakpoint region 1 (EWSR1) is a member of FET (FUS/EWSR1/TAF15) RNA-binding family of proteins. The Ewing sarcoma oncoprotein EWS-FLI1 has been extensively studied, while much less is known about EWSR1 itself, especially the potential role of EWSR1 in response to DNA damage. Here, we found that UV irradiation induces acetylation of EWSR1, which is required for its nucleoli translocation. We identified K423, K432, K438, K640, and K643 as the major acetylation sites, p300/CBP and HDAC3/HDAC10 as the major acetyltransferases and deacetylases, respectively. Mechanically, UV-induced EWSR1 acetylation repressed its interaction with spliceosomal component U1C, which caused abnormal splicing of CHK2, suppressing the activity of CHK2 in response to UV irradiation. Taken together, our findings uncover acetylation as a novel regulatory modification of EWSR1, and is essential for its function in DNA damage response., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.