Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 409
Filter
1.
Nat Commun ; 15(1): 7873, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39251601

ABSTRACT

Meningiomas are associated with inactivation of NF2/Merlin, but approximately one-third of meningiomas with favorable clinical outcomes retain Merlin expression. Biochemical mechanisms underlying Merlin-intact meningioma growth are incompletely understood, and non-invasive biomarkers that may be used to guide treatment de-escalation or imaging surveillance are lacking. Here, we use single-cell RNA sequencing, proximity-labeling proteomic mass spectrometry, mechanistic and functional approaches, and magnetic resonance imaging (MRI) across meningioma xenografts and patients to define biochemical mechanisms and an imaging biomarker that underlie Merlin-intact meningiomas. We find Merlin serine 13 (S13) dephosphorylation drives meningioma Wnt signaling and tumor growth by attenuating inhibitory interactions with ß-catenin and activating the Wnt pathway. MRI analyses show Merlin-intact meningiomas with S13 phosphorylation and favorable clinical outcomes are associated with high apparent diffusion coefficient (ADC). These results define mechanisms underlying a potential imaging biomarker that could be used to guide treatment de-escalation or imaging surveillance for patients with Merlin-intact meningiomas.


Subject(s)
Magnetic Resonance Imaging , Meningeal Neoplasms , Meningioma , Neurofibromin 2 , Wnt Signaling Pathway , Meningioma/diagnostic imaging , Meningioma/metabolism , Meningioma/pathology , Meningioma/genetics , Humans , Phosphorylation , Neurofibromin 2/metabolism , Neurofibromin 2/genetics , Animals , Magnetic Resonance Imaging/methods , Meningeal Neoplasms/diagnostic imaging , Meningeal Neoplasms/metabolism , Meningeal Neoplasms/pathology , Meningeal Neoplasms/genetics , Mice , Cell Line, Tumor , beta Catenin/metabolism , beta Catenin/genetics , Female , Serine/metabolism , Male , Proteomics/methods , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics
2.
Biopreserv Biobank ; 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39321093

ABSTRACT

Introduction: The storage of biospecimens is a substantial source of greenhouse gas emissions and institutional energy costs. Energy-intensive ultra-low temperature (ULT) freezers used for biospecimen storage are a significant source of carbon emissions. ENERGY STAR-certified ULT freezers have the potential to decrease the carbon footprint. Objective: Quantify the impact of an institutional-scale freezer conversion program on carbon emissions and energy costs. Methods: A ULT freezer energy use prediction model was developed to identify and replace the most inefficient freezers in the research building for this pilot, and eventually institution-wide. Multiple linear regression factors included the number of years of use, storage volume, and ENERGY STAR certification status. Electrical usage and carbon emissions were quantified before and after replacement with ENERGY STAR models. Logistical methods were developed to decrease the risks of exposure of frozen samples to ambient temperature during content transfers. Institution-wide energy costs were derived by converting electrical burden to electrical costs. Carbon footprint assessment from ULT freezer operation was computed using the U.S. EPA Greenhouse Gas Equivalencies Calculator. Results: The pilot project revealed an annual reduction of 310,493 kilowatt hours of electrical usage, equivalent to 134 metric tons of carbon emissions. Annual electrical costs were reduced by $55,889 resulting in an 8-year payback on the initial investment. Using the pilot results, we modeled the benefit of the freezer exchange across the entire institution. The modeling predicted that conversion of the institution's remaining 1119 conventional ULT freezers to ENERGY STAR models would lower annual electrical usage by 7,911,549 kilowatt hours (3423 metric tons of carbon emissions), resulting in savings of over $1.4 million annually. Conclusion: Our methods make a large-scale initiative to replace energy-inefficient ULT freezers logistically possible, reduce carbon footprint, and demonstrate an attractive return on investment while proactively protecting valuable research materials.

3.
Neuro Oncol ; 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39248287

ABSTRACT

BACKGROUND: Co-amplification of EGFR and EGFRvIII, a tumor-specific truncation mutant of EGFR, represent hallmark genetic lesions in glioblastoma. METHODS: We used phospho-proteomics, RNA-sequencing, TCGA data and glioblastoma cell culture and mouse models to study the signal transduction mediated by EGFR and EGFRvIII. RESULTS: We report that EGFR and EGFRvIII stimulate the innate immune defense receptor Toll-like Receptor 2 (TLR2); and that knockout of TLR2 dramatically improved survival in orthotopic glioblastoma xenografts. EGFR and EGFRvIII activated TLR2 in a ligand-independent manner, promoting tumor growth and immune evasion. We show that EGFR and EGFRvIII cooperate to activate the Rho-associated protein kinase ROCK2, which modulated malignant progression both by activating TLR2 and WNT signaling, and through remodeling the tumor microenvironment. CONCLUSION: Together, our findings show that EGFR and EGFRvIII cooperate to drive tumor progression through ROCK2 and downstream WNT-ß-catenin/TLR2 signaling pathways.

4.
Nature ; 633(8031): 905-913, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39198643

ABSTRACT

Life-threatening thrombotic events and neurological symptoms are prevalent in COVID-19 and are persistent in patients with long COVID experiencing post-acute sequelae of SARS-CoV-2 infection1-4. Despite the clinical evidence1,5-7, the underlying mechanisms of coagulopathy in COVID-19 and its consequences in inflammation and neuropathology remain poorly understood and treatment options are insufficient. Fibrinogen, the central structural component of blood clots, is abundantly deposited in the lungs and brains of patients with COVID-19, correlates with disease severity and is a predictive biomarker for post-COVID-19 cognitive deficits1,5,8-10. Here we show that fibrin binds to the SARS-CoV-2 spike protein, forming proinflammatory blood clots that drive systemic thromboinflammation and neuropathology in COVID-19. Fibrin, acting through its inflammatory domain, is required for oxidative stress and macrophage activation in the lungs, whereas it suppresses natural killer cells, after SARS-CoV-2 infection. Fibrin promotes neuroinflammation and neuronal loss after infection, as well as innate immune activation in the brain and lungs independently of active infection. A monoclonal antibody targeting the inflammatory fibrin domain provides protection from microglial activation and neuronal injury, as well as from thromboinflammation in the lung after infection. Thus, fibrin drives inflammation and neuropathology in SARS-CoV-2 infection, and fibrin-targeting immunotherapy may represent a therapeutic intervention for patients with acute COVID-19 and long COVID.


Subject(s)
Brain , COVID-19 , Fibrin , Inflammation , Thrombosis , Animals , Female , Humans , Male , Mice , Brain/drug effects , Brain/immunology , Brain/pathology , Brain/virology , COVID-19/immunology , COVID-19/pathology , COVID-19/virology , COVID-19/complications , Fibrin/antagonists & inhibitors , Fibrin/metabolism , Fibrinogen/metabolism , Immunity, Innate , Inflammation/complications , Inflammation/immunology , Inflammation/pathology , Inflammation/virology , Killer Cells, Natural/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Lung/virology , Macrophage Activation/drug effects , Microglia/immunology , Microglia/pathology , Neuroinflammatory Diseases/complications , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/virology , Neurons/pathology , Neurons/virology , Oxidative Stress , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/metabolism , Thrombosis/complications , Thrombosis/immunology , Thrombosis/pathology , Thrombosis/virology , Post-Acute COVID-19 Syndrome/immunology , Post-Acute COVID-19 Syndrome/virology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology
5.
bioRxiv ; 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39149230

ABSTRACT

SARS-CoV-2 continues to pose a threat to public health. Current therapeutics remain limited to direct acting antivirals that lack distinct mechanisms of action and are already showing signs of viral resistance. The virus encodes an ADP-ribosylhydrolase macrodomain (Mac1) that plays an important role in the coronaviral lifecycle by suppressing host innate immune responses. Genetic inactivation of Mac1 abrogates viral replication in vivo by potentiating host innate immune responses. However, it is unknown whether this can be achieved by pharmacologic inhibition and can therefore be exploited therapeutically. Here we report a potent and selective lead small molecule, AVI-4206, that is effective in an in vivo model of SARS-CoV-2 infection. Cellular models indicate that AVI-4206 has high target engagement and can weakly inhibit viral replication in a gamma interferon- and Mac1 catalytic activity-dependent manner; a stronger antiviral effect for AVI-4206 is observed in human airway organoids. In an animal model of severe SARS-CoV-2 infection, AVI-4206 reduces viral replication, potentiates innate immune responses, and leads to a survival benefit. Our results provide pharmacological proof of concept that Mac1 is a valid therapeutic target via a novel immune-restoring mechanism that could potentially synergize with existing therapies targeting distinct, essential aspects of the coronaviral life cycle. This approach could be more widely used to target other viral macrodomains to develop antiviral therapeutics beyond COVID-19.

6.
EMBO Rep ; 25(9): 4078-4090, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39122863

ABSTRACT

The human immunodeficiency virus (HIV-1) is highly dependent on a variety of host factors. Beside proteins, host RNA molecules are reported to aid HIV-1 replication and latency maintenance. Here, we implement multiple workflows of native RNA immunoprecipitation and sequencing (nRIPseq) to determine direct host RNA interaction partners of all 18 HIV-1 (poly)proteins. We identify 1,727 HIV-1 protein - human RNA interactions in the Jurkat cell line and 1,558 interactions in SupT1 cells for a subset of proteins, and discover distinct cellular pathways that seem to be used or controlled by HIV-1 on the RNA level: Tat binds mRNAs of proteins involved in the super elongation complex (AFF1-4, Cyclin-T1). Correlation of the interaction scores (based on binding abundancy) allows identifying the highest confidence interactions, for which we perform a small-scale knockdown screen that leads to the identification of three HIV-1 protein binding RNA interactors involved in HIV-1 replication (AFF2, H4C9 and RPLP0).


Subject(s)
HIV-1 , Proteome , Virus Replication , Humans , HIV-1/genetics , HIV-1/metabolism , HIV-1/physiology , Jurkat Cells , Proteome/metabolism , Virus Replication/genetics , Host-Pathogen Interactions/genetics , Protein Binding , Cyclin T/metabolism , Cyclin T/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , HIV Infections/virology , HIV Infections/metabolism , HIV Infections/genetics , RNA, Viral/metabolism , RNA, Viral/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomal Proteins/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics
7.
Mol Syst Biol ; 20(8): 952-971, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38951684

ABSTRACT

Proximity labeling (PL) via biotinylation coupled with mass spectrometry (MS) captures spatial proteomes in cells. Large-scale processing requires a workflow minimizing hands-on time and enhancing quantitative reproducibility. We introduced a scalable PL pipeline integrating automated enrichment of biotinylated proteins in a 96-well plate format. Combining this with optimized quantitative MS based on data-independent acquisition (DIA), we increased sample throughput and improved protein identification and quantification reproducibility. We applied this pipeline to delineate subcellular proteomes across various compartments. Using the 5HT2A serotonin receptor as a model, we studied temporal changes of proximal interaction networks induced by receptor activation. In addition, we modified the pipeline for reduced sample input to accommodate CRISPR-based gene knockout, assessing dynamics of the 5HT2A network in response to perturbation of selected interactors. This PL approach is universally applicable to PL proteomics using biotinylation-based PL enzymes, enhancing throughput and reproducibility of standard protocols.


Subject(s)
Biotinylation , Proteome , Proteomics , Proteomics/methods , Reproducibility of Results , Humans , Proteome/metabolism , Mass Spectrometry/methods , HEK293 Cells
8.
bioRxiv ; 2024 May 29.
Article in English | MEDLINE | ID: mdl-38854010

ABSTRACT

Genome sequencing efforts have led to the discovery of tens of millions of protein missense variants found in the human population with the majority of these having no annotated role and some likely contributing to trait variation and disease. Sequence-based artificial intelligence approaches have become highly accurate at predicting variants that are detrimental to the function of proteins but they do not inform on mechanisms of disruption. Here we combined sequence and structure-based methods to perform proteome-wide prediction of deleterious variants with information on their impact on protein stability, protein-protein interactions and small-molecule binding pockets. AlphaFold2 structures were used to predict approximately 100,000 small-molecule binding pockets and stability changes for over 200 million variants. To inform on protein-protein interfaces we used AlphaFold2 to predict structures for nearly 500,000 protein complexes. We illustrate the value of mechanism-aware variant effect predictions to study the relation between protein stability and abundance and the structural properties of interfaces underlying trans protein quantitative trait loci (pQTLs). We characterised the distribution of mechanistic impacts of protein variants found in patients and experimentally studied example disease linked variants in FGFR1.

9.
Nat Commun ; 15(1): 4707, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830842

ABSTRACT

Persisting replication intermediates can confer mitotic catastrophe. Loss of the fission yeast telomere protein Taz1 (ortholog of mammalian TRF1/TRF2) causes telomeric replication fork (RF) stalling and consequently, telomere entanglements that stretch between segregating mitotic chromosomes. At ≤20 °C, these entanglements fail to resolve, resulting in lethality. Rif1, a conserved DNA replication/repair protein, hinders the resolution of telomere entanglements without affecting their formation. At mitosis, local nuclear envelope (NE) breakdown occurs in the cell's midregion. Here we demonstrate that entanglement resolution occurs in the cytoplasm following this NE breakdown. However, in response to taz1Δ telomeric entanglements, Rif1 delays midregion NE breakdown at ≤20 °C, in turn disfavoring entanglement resolution. Moreover, Rif1 overexpression in an otherwise wild-type setting causes cold-specific NE defects and lethality, which are rescued by membrane fluidization. Hence, NE properties confer the cold-specificity of taz1Δ lethality, which stems from postponement of NE breakdown. We propose that such postponement promotes clearance of simple stalled RFs, but resolution of complex entanglements (involving strand invasion between nonsister telomeres) requires rapid exposure to the cytoplasm.


Subject(s)
Anaphase , Nuclear Envelope , Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Telomere-Binding Proteins , Telomere , Nuclear Envelope/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Telomere/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces pombe Proteins/genetics , Telomere-Binding Proteins/metabolism , Telomere-Binding Proteins/genetics , DNA Replication
10.
bioRxiv ; 2024 May 24.
Article in English | MEDLINE | ID: mdl-38826258

ABSTRACT

This article describes the Cell Maps for Artificial Intelligence (CM4AI) project and its goals, methods, standards, current datasets, software tools , status, and future directions. CM4AI is the Functional Genomics Data Generation Project in the U.S. National Institute of Health's (NIH) Bridge2AI program. Its overarching mission is to produce ethical, AI-ready datasets of cell architecture, inferred from multimodal data collected for human cell lines, to enable transformative biomedical AI research.

11.
bioRxiv ; 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38712050

ABSTRACT

Chlamydia trachomatis (C.t.), the leading cause of bacterial sexually transmitted infections, employs a type III secretion system (T3SS) to translocate two classes of effectors, inclusion membrane proteins and conventional T3SS (cT3SS) effectors, into the host cell to counter host defense mechanisms. Here we employed three assays to directly evaluate secretion during infection, validating secretion for 23 cT3SS effectors. As bioinformatic analyses have been largely unrevealing, we conducted affinity purification-mass spectrometry to identify host targets and gain insights into the functions of these effectors, identifying high confidence interacting partners for 21 cT3SS effectors. We demonstrate that CebN localizes to the nuclear envelope in infected and bystander cells where it interacts with multiple nucleoporins and Rae1, blocking STAT1 nuclear import following IFN-γ stimulation. By building a cT3SS effector-host interactome, we have identified novel pathways that are targeted during bacterial infection and have begun to address how C.t. effectors combat cell autonomous immunity.

12.
Nat Cancer ; 5(7): 1082-1101, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38816660

ABSTRACT

Dose-limiting toxicity poses a major limitation to the clinical utility of targeted cancer therapies, often arising from target engagement in nonmalignant tissues. This obstacle can be minimized by targeting cancer dependencies driven by proteins with tissue-restricted and/or tumor-restricted expression. In line with another recent report, we show here that, in acute myeloid leukemia (AML), suppression of the myeloid-restricted PIK3CG/p110γ-PIK3R5/p101 axis inhibits protein kinase B/Akt signaling and compromises AML cell fitness. Furthermore, silencing the genes encoding PIK3CG/p110γ or PIK3R5/p101 sensitizes AML cells to established AML therapies. Importantly, we find that existing small-molecule inhibitors against PIK3CG are insufficient to achieve a sustained long-term antileukemic effect. To address this concern, we developed a proteolysis-targeting chimera (PROTAC) heterobifunctional molecule that specifically degrades PIK3CG and potently suppresses AML progression alone and in combination with venetoclax in human AML cell lines, primary samples from patients with AML and syngeneic mouse models.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase , Leukemia, Myeloid, Acute , Proto-Oncogene Proteins c-akt , Signal Transduction , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/genetics , Humans , Animals , Mice , Signal Transduction/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Class Ib Phosphatidylinositol 3-Kinase/genetics , Cell Line, Tumor , Xenograft Model Antitumor Assays , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Proteolysis/drug effects , Female , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
13.
Microbiol Spectr ; 12(7): e0045324, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38814079

ABSTRACT

Chlamydia trachomatis is the leading cause of bacterial sexually transmitted infections in the USA and of preventable blindness worldwide. This obligate intracellular pathogen replicates within a membrane-bound inclusion, but how it acquires nutrients from the host while avoiding detection by the innate immune system is incompletely understood. C. trachomatis accomplishes this in part through the translocation of a unique set of effectors into the inclusion membrane, the inclusion membrane proteins (Incs). Incs are ideally positioned at the host-pathogen interface to reprogram host signaling by redirecting proteins or organelles to the inclusion. Using a combination of co-affinity purification, immunofluorescence confocal imaging, and proteomics, we characterize the interaction between an early-expressed Inc of unknown function, Tri1, and tumor necrosis factor receptor-associated factor 7 (TRAF7). TRAF7 is a multi-domain protein with a RING finger ubiquitin ligase domain and a C-terminal WD40 domain. TRAF7 regulates several innate immune signaling pathways associated with C. trachomatis infection and is mutated in a subset of tumors. We demonstrate that Tri1 and TRAF7 specifically interact during infection and that TRAF7 is recruited to the inclusion. We further show that the predicted coiled-coil domain of Tri1 is necessary to interact with the TRAF7 WD40 domain. Finally, we demonstrate that Tri1 displaces the native TRAF7 binding partners, mitogen-activated protein kinase kinase kinase 2 (MEKK2), and MEKK3. Together, our results suggest that by displacing TRAF7 native binding partners, Tri1 has the capacity to alter TRAF7 signaling during C. trachomatis infection.IMPORTANCEChlamydia trachomatis is the leading cause of bacterial sexually transmitted infections in the USA and preventable blindness worldwide. Although easily treated with antibiotics, the vast majority of infections are asymptomatic and therefore go untreated, leading to infertility and blindness. This obligate intracellular pathogen evades the immune response, which contributes to these outcomes. Here, we characterize the interaction between a C. trachomatis-secreted effector, Tri1, and a host protein involved in innate immune signaling, TRAF7. We identified host proteins that bind to TRAF7 and demonstrated that Tri1 can displace these proteins upon binding to TRAF7. Remarkably, the region of TRAF7 to which these host proteins bind is often mutated in a subset of human tumors. Our work suggests a mechanism by which Tri1 may alter TRAF7 signaling and has implications not only in the pathogenesis of C. trachomatis infections but also in understanding the role of TRAF7 in cancer.


Subject(s)
Bacterial Proteins , Chlamydia Infections , Chlamydia trachomatis , Host-Pathogen Interactions , Humans , Chlamydia trachomatis/metabolism , Chlamydia trachomatis/genetics , Chlamydia trachomatis/immunology , HeLa Cells , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Chlamydia Infections/microbiology , Chlamydia Infections/metabolism , Chlamydia Infections/immunology , Signal Transduction , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Immunity, Innate , Protein Binding , Membrane Proteins/metabolism , Membrane Proteins/genetics , HEK293 Cells
14.
ArXiv ; 2024 May 15.
Article in English | MEDLINE | ID: mdl-38800652

ABSTRACT

Proteins congregate into complexes to perform fundamental cellular functions. Phenotypic outcomes, in health and disease, are often mechanistically driven by the remodeling of protein complexes by protein-coding mutations or cellular signaling changes in response to molecular cues. Here, we present an affinity purification-mass spectrometry (APMS) proteomics protocol to quantify and visualize global changes in protein-protein interaction (PPI) networks between pairwise conditions. We describe steps for expressing affinity-tagged "bait" proteins in mammalian cells, identifying purified protein complexes, quantifying differential PPIs, and visualizing differential PPI networks. Specifically, this protocol details steps for designing affinity-tagged "bait" gene constructs, transfection, affinity purification, mass spectrometry sample preparation, data acquisition, database search, data quality control, PPI confidence scoring, cross-run normalization, statistical data analysis, and differential PPI visualization. Our protocol discusses caveats and limitations with applicability across cell types and biological areas. For complete details on the use and execution of this protocol, please refer to Bouhaddou et al. 20231.

15.
Traffic ; 25(4): e12933, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38600522

ABSTRACT

Macroautophagy/autophagy is an essential catabolic process that targets a wide variety of cellular components including proteins, organelles, and pathogens. ATG7, a protein involved in the autophagy process, plays a crucial role in maintaining cellular homeostasis and can contribute to the development of diseases such as cancer. ATG7 initiates autophagy by facilitating the lipidation of the ATG8 proteins in the growing autophagosome membrane. The noncanonical isoform ATG7(2) is unable to perform ATG8 lipidation; however, its cellular regulation and function are unknown. Here, we uncovered a distinct regulation and function of ATG7(2) in contrast with ATG7(1), the canonical isoform. First, affinity-purification mass spectrometry analysis revealed that ATG7(2) establishes direct protein-protein interactions (PPIs) with metabolic proteins, whereas ATG7(1) primarily interacts with autophagy machinery proteins. Furthermore, we identified that ATG7(2) mediates a decrease in metabolic activity, highlighting a novel splice-dependent function of this important autophagy protein. Then, we found a divergent expression pattern of ATG7(1) and ATG7(2) across human tissues. Conclusively, our work uncovers the divergent patterns of expression, protein interactions, and function of ATG7(2) in contrast to ATG7(1). These findings suggest a molecular switch between main catabolic processes through isoform-dependent expression of a key autophagy gene.


Subject(s)
Autophagy , Energy Metabolism , Humans , Autophagosomes/metabolism , Autophagy-Related Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Protein Isoforms/metabolism
16.
G3 (Bethesda) ; 14(7)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38657142

ABSTRACT

In fission yeast lacking the telomere binding protein, Taz1, replication forks stall at telomeres, triggering deleterious downstream events. Strand invasion from one taz1Δ telomeric stalled fork to another on a separate (nonsister) chromosome leads to telomere entanglements, which are resolved in mitosis at 32°C; however, entanglement resolution fails at ≤20°C, leading to cold-specific lethality. Previously, we found that loss of the mitotic function of Rif1, a conserved DNA replication and repair factor, suppresses cold sensitivity by promoting resolution of entanglements without affecting entanglement formation. To understand the underlying pathways of mitotic entanglement resolution, we performed a series of genome-wide synthetic genetic array screens to generate a comprehensive list of genetic interactors of taz1Δ and rif1Δ. We modified a previously described screening method to ensure that the queried cells were kept in log phase growth. In addition to recapitulating previously identified genetic interactions, we find that loss of genes encoding components of the nuclear pore complex (NPC) promotes telomere disentanglement and suppresses taz1Δ cold sensitivity. We attribute this to more rapid anaphase midregion nuclear envelope (NE) breakdown in the absence of these NPC components. Loss of genes involved in lipid metabolism reverses the ability of rif1+ deletion to suppress taz1Δ cold sensitivity, again pinpointing NE modulation. A rif1+ separation-of-function mutant that specifically loses Rif1's mitotic functions yields similar genetic interactions. Genes promoting membrane fluidity were enriched in a parallel taz1+ synthetic lethal screen at permissive temperature, cementing the idea that the cold specificity of taz1Δ lethality stems from altered NE homeostasis.


Subject(s)
Homeostasis , Nuclear Envelope , Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Telomere-Binding Proteins , Telomere , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Telomere/genetics , Telomere/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Nuclear Envelope/metabolism , Nuclear Envelope/genetics , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Mitosis/genetics , Genetic Testing , Nuclear Pore/metabolism , Nuclear Pore/genetics
17.
Nat Chem Biol ; 20(9): 1133-1143, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38528119

ABSTRACT

The µ-opioid receptor (µOR) represents an important target of therapeutic and abused drugs. So far, most understanding of µOR activity has focused on a subset of known signal transducers and regulatory molecules. Yet µOR signaling is coordinated by additional proteins in the interaction network of the activated receptor, which have largely remained invisible given the lack of technologies to interrogate these networks systematically. Here we describe a proteomics and computational approach to map the proximal proteome of the activated µOR and to extract subcellular location, trafficking and functional partners of G-protein-coupled receptor (GPCR) activity. We demonstrate that distinct opioid agonists exert differences in the µOR proximal proteome mediated by endocytosis and endosomal sorting. Moreover, we identify two new µOR network components, EYA4 and KCTD12, which are recruited on the basis of receptor-triggered G-protein activation and might form a previously unrecognized buffering system for G-protein activity broadly modulating cellular GPCR signaling.


Subject(s)
Proteome , Proteomics , Receptors, Opioid, mu , Humans , Endocytosis , HEK293 Cells , Proteome/metabolism , Proteomics/methods , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/agonists , Signal Transduction
18.
bioRxiv ; 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38464023

ABSTRACT

Chlamydia trachomatis is the leading cause of bacterial sexually transmitted infections in the US and of preventable blindness worldwide. This obligate intracellular pathogen replicates within a membrane-bound inclusion, but how it acquires nutrients from the host while avoiding detection by the innate immune system is incompletely understood. C. trachomatis accomplishes this in part through the translocation of a unique set of effectors into the inclusion membrane, the inc lusion membrane proteins (Incs). Incs are ideally positioned at the host-pathogen interface to reprogram host signaling by redirecting proteins or organelles to the inclusion. Using a combination of co-affinity purification, immunofluorescence confocal imaging, and proteomics, we characterize the interaction between an early-expressed Inc of unknown function, Tri1, and tumor necrosis factor receptor associated factor 7 (TRAF7). TRAF7 is a multi-domain protein with a RING finger ubiquitin ligase domain and a C-terminal WD40 domain. TRAF7 regulates several innate immune signaling pathways associated with C. trachomatis infection and is mutated in a subset of tumors. We demonstrate that Tri1 and TRAF7 specifically interact during infection and that TRAF7 is recruited to the inclusion. We further show that the predicted coiled-coil domain of Tri1 is necessary to interact with the TRAF7 WD40 domain. Finally, we demonstrate that Tri1 displaces the native TRAF7 binding partners, mitogen activated protein kinase kinase kinase 2 (MEKK2) and MEKK3. Together, our results suggest that by displacing TRAF7 native binding partners, Tri1 has the capacity to alter TRAF7 signaling during C. trachomatis infection. Importance: Chlamydia trachomatis is the leading cause of bacterial sexually transmitted infections in the US and preventable blindness worldwide. Although easily treated with antibiotics, the vast majority of infections are asymptomatic and therefore go untreated, leading to infertility and blindness. This obligate intracellular pathogen evades the immune response, which contributes to these outcomes. Here, we characterize the interaction between a C. trachomatis secreted effector, Tri1, and a host protein involved in innate immune signaling, TRAF7. We identified host proteins that bind to TRAF7 and demonstrate that Tri1 can displace these proteins upon binding to TRAF7. Remarkably, the region of TRAF7 to which these host proteins bind is often mutated in a subset of human tumors. Our work suggests a mechanism by which Tri1 may alter TRAF7 signaling and has implications not only in the pathogenesis of C. trachomatis infections, but also in understanding the role of TRAF7 in cancer.

19.
bioRxiv ; 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38464290

ABSTRACT

Sensory signaling pathways use adaptation to dynamically respond to changes in their environment. Here, we report the mechanism of sensory adaptation in the Pil-Chp mechanosensory system, which the important human pathogen Pseudomonas aeruginosa uses to sense mechanical stimuli during surface exploration. Using biochemistry, genetics, and cell biology, we discovered that the enzymes responsible for adaptation, a methyltransferase and a methylesterase, are segregated to opposing cell poles as P. aeruginosa explore surfaces. By coordinating the localization of both enzymes, we found that the Pil-Chp response regulators influence local receptor methylation, the molecular basis of bacterial sensory adaptation. We propose a model in which adaptation during mechanosensing spatially resets local receptor methylation, and thus Pil-Chp signaling, to modulate the pathway outputs, which are involved in P. aeruginosa virulence. Despite decades of bacterial sensory adaptation studies, our work has uncovered an unrecognized mechanism that bacteria use to achieve adaptation to sensory stimuli.

20.
Mol Cell Proteomics ; 23(5): 100755, 2024 May.
Article in English | MEDLINE | ID: mdl-38548018

ABSTRACT

Human APOBEC3 enzymes are a family of single-stranded (ss)DNA and RNA cytidine deaminases that act as part of the intrinsic immunity against viruses and retroelements. These enzymes deaminate cytosine to form uracil which can functionally inactivate or cause degradation of viral or retroelement genomes. In addition, APOBEC3s have deamination-independent antiviral activity through protein and nucleic acid interactions. If expression levels are misregulated, some APOBEC3 enzymes can access the human genome leading to deamination and mutagenesis, contributing to cancer initiation and evolution. While APOBEC3 enzymes are known to interact with large ribonucleoprotein complexes, the function and RNA dependence are not entirely understood. To further understand their cellular roles, we determined by affinity purification mass spectrometry (AP-MS) the protein interaction network for the human APOBEC3 enzymes and mapped a diverse set of protein-protein and protein-RNA mediated interactions. Our analysis identified novel RNA-mediated interactions between APOBEC3C, APOBEC3H Haplotype I and II, and APOBEC3G with spliceosome proteins, and APOBEC3G and APOBEC3H Haplotype I with proteins involved in tRNA methylation and ncRNA export from the nucleus. In addition, we identified RNA-independent protein-protein interactions with APOBEC3B, APOBEC3D, and APOBEC3F and the prefoldin family of protein-folding chaperones. Interaction between prefoldin 5 (PFD5) and APOBEC3B disrupted the ability of PFD5 to induce degradation of the oncogene cMyc, implicating the APOBEC3B protein interaction network in cancer. Altogether, the results uncover novel functions and interactions of the APOBEC3 family and suggest they may have fundamental roles in cellular RNA biology, their protein-protein interactions are not redundant, and there are protein-protein interactions with tumor suppressors, suggesting a role in cancer biology. Data are available via ProteomeXchange with the identifier PXD044275.


Subject(s)
Cytidine Deaminase , Protein Interaction Maps , Humans , Cytidine Deaminase/metabolism , Cytidine Deaminase/genetics , Deamination , APOBEC Deaminases/metabolism , Aminohydrolases/metabolism , Aminohydrolases/genetics , HEK293 Cells , Cytosine Deaminase/metabolism , APOBEC-3G Deaminase/metabolism , APOBEC-3G Deaminase/genetics , Spliceosomes/metabolism , Protein Binding , Mass Spectrometry , RNA/metabolism , Minor Histocompatibility Antigens/metabolism , Minor Histocompatibility Antigens/genetics
SELECTION OF CITATIONS
SEARCH DETAIL