Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45.959
Filter
1.
Sci Adv ; 10(31): eaax2323, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39093972

ABSTRACT

The nucleocapsid protein of severe acute respiratory syndrome coronavirus 2 encapsidates the viral genome and is essential for viral function. The central disordered domain comprises a serine-arginine-rich (SR) region that is hyperphosphorylated in infected cells. This modification regulates function, although mechanistic details remain unknown. We use nuclear magnetic resonance to follow structural changes occurring during hyperphosphorylation by serine arginine protein kinase 1, glycogen synthase kinase 3, and casein kinase 1, that abolishes interaction with RNA. When eight approximately uniformly distributed sites have been phosphorylated, the SR domain binds the same interface as single-stranded RNA, resulting in complete inhibition of RNA binding. Phosphorylation by protein kinase A does not prevent RNA binding, indicating that the pattern resulting from physiologically relevant kinases is specific for inhibition. Long-range contacts between the RNA binding, linker, and dimerization domains are abrogated, phenomena possibly related to genome packaging and unpackaging. This study provides insight into the recruitment of specific host kinases to regulate viral function.


Subject(s)
Coronavirus Nucleocapsid Proteins , Protein Binding , RNA, Viral , SARS-CoV-2 , Phosphorylation , SARS-CoV-2/metabolism , Coronavirus Nucleocapsid Proteins/metabolism , Coronavirus Nucleocapsid Proteins/chemistry , Humans , RNA, Viral/metabolism , RNA, Viral/chemistry , Protein Conformation , COVID-19/virology , COVID-19/metabolism , Nucleocapsid Proteins/metabolism , Nucleocapsid Proteins/chemistry , Models, Molecular , Binding Sites , Phosphoproteins
2.
Nat Commun ; 15(1): 7016, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39147754

ABSTRACT

Owing to its roles in cellular signal transduction, protein phosphorylation plays critical roles in myriad cell processes. That said, detecting and quantifying protein phosphorylation has remained a challenge. We describe the use of a novel mass spectrometer (Orbitrap Astral) coupled with data-independent acquisition (DIA) to achieve rapid and deep analysis of human and mouse phosphoproteomes. With this method, we map approximately 30,000 unique human phosphorylation sites within a half-hour of data collection. The technology is benchmarked to other state-of-the-art MS platforms using both synthetic peptide standards and with EGF-stimulated HeLa cells. We apply this approach to generate a phosphoproteome multi-tissue atlas of the mouse. Altogether, we detect 81,120 unique phosphorylation sites within 12 hours of measurement. With this unique dataset, we examine the sequence, structural, and kinase specificity context of protein phosphorylation. Finally, we highlight the discovery potential of this resource with multiple examples of phosphorylation events relevant to mitochondrial and brain biology.


Subject(s)
Mass Spectrometry , Phosphoproteins , Proteome , Proteomics , Humans , Phosphoproteins/metabolism , Phosphoproteins/analysis , Animals , HeLa Cells , Phosphorylation , Mice , Proteome/metabolism , Mass Spectrometry/methods , Proteomics/methods
3.
Anal Chem ; 96(33): 13358-13370, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39102789

ABSTRACT

Formalin-fixed paraffin-embedded (FFPE) tissues are suitable for proteomic and phosphoproteomic biomarker studies by data-independent acquisition mass spectrometry. The choice of the sample preparation method influences the number, intensity, and reproducibility of identifications. By comparing four deparaffinization and rehydration methods, including heptane, histolene, SubX, and xylene, we found that heptane and methanol produced the lowest coefficients of variation (CVs). Using this, five extraction methods from the literature were modified and evaluated for their performance using kidney, leg muscle, lung, and testicular rat organs. All methods performed well, except for SP3 due to insufficient tissue lysis. Heat n' Beat was the fastest and most reproducible method with the highest digestion efficiency and lowest CVs. S-Trap produced the highest peptide yield, while TFE produced the best phosphopeptide enrichment efficiency. The quantitation of FFPE-derived peptides remains an ongoing challenge with bias in UV and fluorescence assays across methods, most notably in SPEED. Functional enrichment analysis demonstrated that each method favored extracting some gene ontology cellular components over others including chromosome, cytoplasmic, cytoskeleton, endoplasmic reticulum, membrane, mitochondrion, and nucleoplasm protein groups. The outcome is a set of recommendations for choosing the most appropriate method for different settings.


Subject(s)
Paraffin Embedding , Proteomics , Proteomics/methods , Animals , Rats , Formaldehyde/chemistry , Male , Phosphoproteins/analysis , Phosphoproteins/metabolism , Phosphoproteins/isolation & purification , Tissue Fixation , Kidney/metabolism , Kidney/chemistry
4.
Commun Biol ; 7(1): 974, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39127848

ABSTRACT

Calorie restriction (CR) and treatment with rapamycin (RM), an inhibitor of the mTORC1 growth-promoting signaling pathway, are known to slow aging and promote health from worms to humans. At the transcriptome and proteome levels, long-term CR and RM treatments have partially overlapping effects, while their impact on protein phosphorylation within cellular signaling pathways have not been compared. Here we measured the phosphoproteomes of soleus, tibialis anterior, triceps brachii and gastrocnemius muscles from adult (10 months) and 30-month-old (aged) mice receiving either a control, a calorie restricted or an RM containing diet from 15 months of age. We reproducibly detected and extensively analyzed a total of 6960 phosphosites, 1415 of which are not represented in standard repositories. We reveal the effect of these interventions on known mTORC1 pathway substrates, with CR displaying greater between-muscle variation than RM. Overall, CR and RM have largely consistent, but quantitatively distinct long-term effects on the phosphoproteome, mitigating age-related changes to different degrees. Our data expands the catalog of protein phosphorylation sites in the mouse, providing important information regarding their tissue-specificity, and revealing the impact of long-term nutrient-sensing pathway inhibition on mouse skeletal muscle.


Subject(s)
Aging , Caloric Restriction , Muscle, Skeletal , Sirolimus , Animals , Phosphorylation , Aging/metabolism , Sirolimus/pharmacology , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Male , Mice, Inbred C57BL , Mechanistic Target of Rapamycin Complex 1/metabolism , Signal Transduction/drug effects , Proteome/metabolism , Phosphoproteins/metabolism , Phosphoproteins/genetics , Muscle Proteins/metabolism
5.
Front Immunol ; 15: 1439184, 2024.
Article in English | MEDLINE | ID: mdl-39104541

ABSTRACT

Introduction: Human Cytomegalovirus (HCMV) is a betaherpesvirus that causes severe disease in immunocompromised transplant recipients. Immunotherapy with CD8 T cells specific for HCMV antigens presented on HLA class-I molecules is explored as strategy for long-term relief to such patients, but the antiviral effectiveness of T cell preparations cannot be efficiently predicted by available methods. Methods: We developed an Assay for Rapid Measurement of Antiviral T-cell Activity (ARMATA) by real-time automated fluorescent microscopy and used it to study the ability of CD8 T cells to neutralize HCMV and control its spread. As a proof of principle, we used TCR-transgenic T cells specific for the immunodominant HLA-A02-restricted tegumental phosphoprotein pp65. pp65 expression follows an early/late kinetic, but it is not clear at which stage of the virus cycle it acts as an antigen. We measured control of HCMV infection by T cells as early as 6 hours post infection (hpi). Results: The timing of the antigen recognition indicated that it occurred before the late phase of the virus cycle, but also that virion-associated pp65 was not recognized during virus entry into cells. Monitoring of pp65 gene expression dynamics by reporter fluorescent genes revealed that pp65 was detectable as early as 6 hpi, and that a second and much larger bout of expression occurs in the late phase of the virus cycle by 48 hpi. Since transgenic (Tg)-pp65 specific CD8 T cells were activated even when DNA replication was blocked, our data argue that pp65 acts as an early virus gene for immunological purposes. Discussion: ARMATA does not only allow same day identification of antiviral T-cell activity, but also provides a method to define the timing of antigen recognition in the context of HCMV infection.


Subject(s)
CD8-Positive T-Lymphocytes , Cytomegalovirus Infections , Cytomegalovirus , Phosphoproteins , Viral Matrix Proteins , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus/immunology , Cytomegalovirus/genetics , Phosphoproteins/immunology , Phosphoproteins/genetics , Humans , Viral Matrix Proteins/immunology , Viral Matrix Proteins/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Gene Expression Regulation, Viral , Antigens, Viral/immunology , HLA-A2 Antigen/immunology , HLA-A2 Antigen/genetics
6.
Immun Inflamm Dis ; 12(8): e1367, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39119967

ABSTRACT

BACKGROUND: Retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations (RVCL-S) is a rare autosomal dominant systemic microvascular disorder attributed to TREX1 (three-prime repair exonuclease-1) gene mutations, often proned to misdiagnosed. METHODS: We reported a case of RVCL-S coexisting with systemic lupus erythematosus due to a mutation in the TREX1 gene. This study provided a summary and discussion of previously documented cases related to TREX1 mutations or RVCL-S. RESULTS: A 39-year-old female patient visited the clinic due to progressive memory loss and speech difficulties. Magnetic resonance imaging results showed corpus callosum atrophy and multiple subcortical calcifications in both brain hemispheres. Genetic testing revealed a TREX1 gene mutation (c.294dupA). Treatment with immunosuppressive therapy for 2 months led to improvements in communication and mobility. We also summarized previously reported cases providing an overview of TREX1 gene mutation or RCVL-S. CONCLUSION: Our case establishes a compelling foundation for future RVCL-S diagnosis and treatment paradigms. Notably, conducting systemic immunity screening in patients with RVCL-S emerges as a strategic approach to prevent potential diagnostic oversights.


Subject(s)
Exodeoxyribonucleases , Leukoencephalopathies , Lupus Erythematosus, Systemic , Mutation , Humans , Female , Adult , Exodeoxyribonucleases/genetics , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/genetics , Leukoencephalopathies/diagnosis , Leukoencephalopathies/genetics , Leukoencephalopathies/etiology , Phosphoproteins/genetics , Diagnostic Errors/prevention & control , Magnetic Resonance Imaging , Retinal Vasculitis/diagnosis , Retinal Vasculitis/etiology , Retinal Diseases , Vascular Diseases , Hereditary Central Nervous System Demyelinating Diseases
7.
Cells ; 13(15)2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39120297

ABSTRACT

Nucleophosmin (NPM1) is a key nucleolar protein released from the nucleolus in response to stress stimuli. NPM1 functions as a stress regulator with nucleic acid and protein chaperone activities, rapidly shuttling between the nucleus and cytoplasm. NPM1 is ubiquitously expressed in tissues and can be found in the nucleolus, nucleoplasm, cytoplasm, and extracellular environment. It plays a central role in various biological processes such as ribosome biogenesis, cell cycle regulation, cell proliferation, DNA damage repair, and apoptosis. In addition, it is highly expressed in cancer cells and solid tumors, and its mutation is a major cause of acute myeloid leukemia (AML). This review focuses on NPM1's structural features, functional diversity, subcellular distribution, and role in stress modulation.


Subject(s)
Cell Nucleolus , Nuclear Proteins , Nucleophosmin , Stress, Physiological , Humans , Nuclear Proteins/metabolism , Cell Nucleolus/metabolism , Animals , Phosphoproteins/metabolism
8.
Int J Mol Sci ; 25(15)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39125875

ABSTRACT

Parasites have been associated with possible anticancer activity, including Trypanosoma cruzi, which has been linked to inhibiting the growth of solid tumors. To better understand this antitumor effect, we investigated the association of anti-T. cruzi antibodies with B cells of the acute lymphoblastic leukemia (ALL) SUPB15 cell line. The antibodies were generated in rabbits. IgGs were purified by affinity chromatography. Two procedures (flow cytometry (CF) and Western blot(WB)) were employed to recognize anti-T. cruzi antibodies on SUPB15 cells. We also used CF to determine whether the anti-T. cruzi antibodies could suppress SUPB15 cells. The anti-T. cruzi antibodies recognized 35.5% of the surface antigens of SUPB15. The complement-dependent cytotoxicity (CDC) results demonstrate the cross-suppression of anti-T. cruzi antibodies on up to 8.4% of SUPB15 cells. For the WB analysis, a band at 100 kDa with high intensity was sequenced using mass spectrometry, identifying the protein as nucleolin. This protein may play a role in the antitumor effect on T. cruzi. The anti-T. cruzi antibodies represent promising polyclonal antibodies that have the effect of tumor-suppressive cross-linking on cancer cells, which should be further investigated.


Subject(s)
Antibodies, Protozoan , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Trypanosoma cruzi , Trypanosoma cruzi/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Humans , Cell Line, Tumor , Animals , Rabbits , Antibodies, Protozoan/immunology , RNA-Binding Proteins/immunology , RNA-Binding Proteins/metabolism , Nucleolin , Phosphoproteins/immunology , Phosphoproteins/metabolism
9.
Nat Commun ; 15(1): 6777, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117624

ABSTRACT

Metabolic rewiring during the proliferation-to-quiescence transition is poorly understood. Here, using a model of contact inhibition-induced quiescence, we conducted 13C-metabolic flux analysis in proliferating (P) and quiescent (Q) mouse embryonic fibroblasts (MEFs) to investigate this process. Q cells exhibit reduced glycolysis but increased TCA cycle flux and mitochondrial respiration. Reduced glycolytic flux in Q cells correlates with reduced glycolytic enzyme expression mediated by yes-associated protein (YAP) inhibition. The increased TCA cycle activity and respiration in Q cells is mediated by induced mitochondrial pyruvate carrier (MPC) expression, rendering them vulnerable to MPC inhibition. The malate-to-pyruvate flux, which generates NADPH, is markedly reduced by modulating malic enzyme 1 (ME1) dimerization in Q cells. Conversely, the malate dehydrogenase 1 (MDH1)-mediated oxaloacetate-to-malate flux is reversed and elevated in Q cells, driven by high mitochondrial-derived malate levels, reduced cytosolic oxaloacetate, elevated MDH1 levels, and a high cytoplasmic NAD+/NADH ratio. Transcriptomic analysis revealed large number of genes are induced in Q cells, many of which are associated with the extracellular matrix (ECM), while YAP-dependent and cell cycle-related genes are repressed. The results suggest that high TCA cycle flux and respiration in Q cells are required to generate ATP and amino acids to maintain de-novo ECM protein synthesis and secretion.


Subject(s)
Adaptor Proteins, Signal Transducing , Citric Acid Cycle , Contact Inhibition , Fibroblasts , Glycolysis , Malate Dehydrogenase , Mitochondria , Transcriptome , YAP-Signaling Proteins , Animals , YAP-Signaling Proteins/metabolism , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Fibroblasts/metabolism , Malate Dehydrogenase/metabolism , Malate Dehydrogenase/genetics , Mitochondria/metabolism , Malates/metabolism , Cell Proliferation , Pyruvic Acid/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Phosphoproteins/metabolism , Phosphoproteins/genetics
10.
Virol J ; 21(1): 186, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39135075

ABSTRACT

BACKGROUND: The global outbreak of COVID-19 caused by the SARS-CoV-2 has led to millions of deaths. This unanticipated emergency has prompted virologists across the globe to delve deeper into the intricate dynamicity of the host-virus interface with an aim to identify antiviral targets and elucidate host and viral determinants of severe disease. AIM: The present study was undertaken to analyse the role of histone deacetylase 6 (HDAC6) in regulating SARS-CoV-2 infection. RESULTS: Gradual increase in HDAC6 expression was observed in different SARS-CoV-2-permissive cell lines following SARS-CoV-2 infection. The SARS-CoV-2 nucleocapsid protein (N protein) was identified as the primary viral factor responsible for upregulating HDAC6 expression. Downregulation of HDAC6 using shRNA or a specific inhibitor tubacin resulted in reduced viral replication suggesting proviral role of its deacetylase activity. Further investigations uncovered the interaction of HDAC6 with stress granule protein G3BP1 and N protein during infection. HDAC6-mediated deacetylation of SARS-CoV-2 N protein was found to be crucial for its association with G3BP1. CONCLUSION: This study provides valuable insights into the molecular mechanisms underlying the disruption of cytoplasmic stress granules during SARS-CoV-2 infection and highlights the significance of HDAC6 in the process.


Subject(s)
COVID-19 , Coronavirus Nucleocapsid Proteins , Histone Deacetylase 6 , SARS-CoV-2 , Virus Replication , Histone Deacetylase 6/metabolism , Histone Deacetylase 6/genetics , Humans , SARS-CoV-2/physiology , Coronavirus Nucleocapsid Proteins/metabolism , Coronavirus Nucleocapsid Proteins/genetics , COVID-19/virology , COVID-19/metabolism , RNA Recognition Motif Proteins/metabolism , Acetylation , Cell Line , Chlorocebus aethiops , Phosphoproteins/metabolism , Phosphoproteins/genetics , Vero Cells , Animals , Host-Pathogen Interactions , Poly-ADP-Ribose Binding Proteins , DNA Helicases , RNA Helicases
11.
Sci Rep ; 14(1): 19496, 2024 08 22.
Article in English | MEDLINE | ID: mdl-39174635

ABSTRACT

Anaplastic thyroid carcinoma (ATC) is a highly aggressive human malignancy without effective treatment. Yes-associated protein (YAP) is a critical effector of the Hippo pathway, which is essential in thyroid carcinogenesis. However, the underlying mechanisms of aberrant YAP expression in ATC are not completely understood. Ubiquitylation-related enzyme siRNA screening identified the ubiquitin protein ligase E3 component n-recognin 1 (UBR1) as a stabilizer of YAP in ATC cells. UBR1 deficiency reduced YAP protein levels and its target gene expression. UBR1 directly interacted with YAP and promoted its monoubiquitylation, competitively suppressing its polyubiquitylation and resulting in extended protein half-life. UBR1 depletion reduced ATC cell proliferation and migration in vitro. Xenograft tumor studies also suggested that UBR1 knockdown suppressed ATC cell growth in vivo. Furthermore, exogenous YAP expression partially reversed the inhibitive effects of UBR1 depletion on ATC cell proliferation and migration. Our studies demonstrated that UBR1 directly interacts with YAP and stabilized it in a monoubiquitylation-dependent manner, consequently promoting ATC tumorigenesis, suggesting that UBR1 might be a potentially therapeutic target for ATC treatment.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Movement , Cell Proliferation , Thyroid Carcinoma, Anaplastic , Transcription Factors , Ubiquitination , YAP-Signaling Proteins , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Thyroid Carcinoma, Anaplastic/metabolism , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Carcinoma, Anaplastic/genetics , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Mice , Protein Stability , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Thyroid Neoplasms/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Disease Progression , Mice, Nude , Gene Expression Regulation, Neoplastic , Phosphoproteins/metabolism , Phosphoproteins/genetics
12.
Sci Rep ; 14(1): 19479, 2024 08 22.
Article in English | MEDLINE | ID: mdl-39174793

ABSTRACT

Genes of the Sprouty family (Spry1-4) are feedback inhibitors of receptor tyrosine kinases, especially of Ret and the FGF receptors. As such, they play distinct and overlapping roles in embryo morphogenesis and are considered to be tumor suppressors in adult life. Genetic experiments in mice have defined in great detail the role of these genes during embryonic development, however their function in adult mice is less clearly established. Here we generate adult-onset, whole body Spry1/2/4 triple knockout mice. Tumor incidence in triple mutant mice is comparable to that of wild type littermates of up to one year of age, indicating that Sprouty loss per se is not sufficient to initiate tumorigenesis. On the other hand, triple knockout mice do not gain weight as they age, show less visceral fat, and have lower plasma glucose levels than wild type littermates, despite showing similar food intake and slightly reduced motor function. They also show alopecia, eyelid inflammation, and mild hyperthyroidism. Finally, triple knockout mice present phosphaturia and hypophosphatemia, suggesting exacerbated signaling downstream of FGF23. In conclusion, triple knockout mice develop a series of endocrine abnormalities but do not show increased tumor incidence.


Subject(s)
Adaptor Proteins, Signal Transducing , Fibroblast Growth Factor-23 , Membrane Proteins , Mice, Knockout , Phosphoproteins , Animals , Mice , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Intracellular Signaling Peptides and Proteins/genetics , Female , Male , Endocrine System Diseases/genetics , Endocrine System Diseases/metabolism , Nerve Tissue Proteins , Protein Serine-Threonine Kinases
13.
Cell Mol Life Sci ; 81(1): 339, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39120648

ABSTRACT

Senataxin is an evolutionarily conserved DNA/RNA helicase, whose dysfunctions are linked to neurodegeneration and cancer. A main activity of this protein is the removal of R-loops, which are nucleic acid structures capable to promote DNA damage and replication stress. Here we found that Senataxin deficiency causes the release of damaged DNA into extranuclear bodies, called micronuclei, triggering the massive recruitment of cGAS, the apical sensor of the innate immunity pathway, and the downstream stimulation of interferon genes. Such cGAS-positive micronuclei are characterized by defective membrane envelope and are particularly abundant in cycling cells lacking Senataxin, but not after exposure to a DNA breaking agent or in absence of the tumor suppressor BRCA1 protein, a partner of Senataxin in R-loop removal. Micronuclei with a discontinuous membrane are normally cleared by autophagy, a process that we show is impaired in Senataxin-deficient cells. The formation of Senataxin-dependent inflamed micronuclei is promoted by the persistence of nuclear R-loops stimulated by the DSIF transcription elongation complex and the engagement of EXO1 nuclease activity on nuclear DNA. Coherently, high levels of EXO1 result in poor prognosis in a subset of tumors lacking Senataxin expression. Hence, R-loop homeostasis impairment, together with autophagy failure and unscheduled EXO1 activity, elicits innate immune response through micronuclei formation in cells lacking Senataxin.


Subject(s)
Autophagy , DNA Damage , DNA Helicases , Inflammation , Multifunctional Enzymes , Nucleotidyltransferases , R-Loop Structures , RNA Helicases , Humans , Autophagy/genetics , BRCA1 Protein/metabolism , BRCA1 Protein/genetics , BRCA1 Protein/deficiency , DNA Helicases/metabolism , DNA Helicases/genetics , DNA Helicases/deficiency , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/deficiency , Exodeoxyribonucleases/metabolism , Exodeoxyribonucleases/genetics , Immunity, Innate , Inflammation/pathology , Inflammation/metabolism , Inflammation/genetics , Multifunctional Enzymes/metabolism , Multifunctional Enzymes/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Phosphoproteins , RNA Helicases/metabolism , RNA Helicases/genetics
14.
Retina ; 44(9): 1580-1589, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39167579

ABSTRACT

PURPOSE: To determine the association between gene-expression profiling (GEP), next-generation sequencing (NGS), preferentially expressed antigen in melanoma (PRAME) features, and metastatic risk in patients with uveal melanoma (UM). METHODS: A retrospective analysis of patients with UM treated by brachytherapy or enucleation by a single ocular oncologist was conducted from November 2020 and July 2022. Clinicopathologic features, patient outcomes, GEP classification, NGS, and PRAME results were recorded. RESULTS: Comprehensive GEP, PRAME, and NGS testing was performed on 135 UMs. The presence of eukaryotic translation initiation factor 1A, X-chromosomal and splicing factor 3B subunit 1 mutations was significantly associated with GEP class 1A and GEP class 1B, respectively. The presence of BRCA- associated protein-1 mutation was significantly associated with GEP class 2. The average largest basal diameter for tumors with eukaryotic translation initiation factor 1A, X-chromosomal mutations was significantly smaller than those with splicing factor 3B subunit 1 mutations and BRCA1-associated protein-1 mutations. Class 2 tumors metastasized sooner than GEP class 1 tumors. Tumors with splicing factor 3B subunit 1 and/or BRCA1-associated protein-1 mutations metastasized sooner compared with tumors that had either no driver mutation or no mutations at all. Tumors with splicing factor 3B subunit 1 did not have a significantly different time to metastasis compared with tumors with BRCA1-associated protein-1 (P value = 0.97). Forty tumors (30%) were PRAME positive, and the remaining 95 tumors (70%) were PRAME negative. Tumors with PRAME-positive status did not have a significantly different time to metastasis compared with tumors without PRAME-positive status (P value = 0.11). CONCLUSION: GEP, NGS, and PRAME expression analysis help determine different levels of metastatic risk in UM. Although other prognostic tests exist, the following study reports on the use of NGS for metastatic prognostication in UM. However, limitations of NGS exist, especially with small lesions that are technically difficult to biopsy.


Subject(s)
Antigens, Neoplasm , Biomarkers, Tumor , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Melanoma , Uveal Neoplasms , Humans , Uveal Neoplasms/genetics , Uveal Neoplasms/diagnosis , Melanoma/genetics , Retrospective Studies , Male , Female , Middle Aged , Antigens, Neoplasm/genetics , Gene Expression Profiling/methods , Aged , Biomarkers, Tumor/genetics , Mutation , Adult , Gene Expression Regulation, Neoplastic , Aged, 80 and over , Eukaryotic Initiation Factor-1/genetics , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Brachytherapy , Phosphoproteins , Tumor Suppressor Proteins , Ubiquitin Thiolesterase
15.
Nature ; 632(8025): 622-629, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39112696

ABSTRACT

Multisystem inflammatory syndrome in children (MIS-C) is a severe, post-infectious sequela of SARS-CoV-2 infection1,2, yet the pathophysiological mechanism connecting the infection to the broad inflammatory syndrome remains unknown. Here we leveraged a large set of samples from patients with MIS-C to identify a distinct set of host proteins targeted by patient autoantibodies including a particular autoreactive epitope within SNX8, a protein involved in regulating an antiviral pathway associated with MIS-C pathogenesis. In parallel, we also probed antibody responses from patients with MIS-C to the complete SARS-CoV-2 proteome and found enriched reactivity against a distinct domain of the SARS-CoV-2 nucleocapsid protein. The immunogenic regions of the viral nucleocapsid and host SNX8 proteins bear remarkable sequence similarity. Consequently, we found that many children with anti-SNX8 autoantibodies also have cross-reactive T cells engaging both the SNX8 and the SARS-CoV-2 nucleocapsid protein epitopes. Together, these findings suggest that patients with MIS-C develop a characteristic immune response to the SARS-CoV-2 nucleocapsid protein that is associated with cross-reactivity to the self-protein SNX8, demonstrating a mechanistic link between the infection and the inflammatory syndrome, with implications for better understanding a range of post-infectious autoinflammatory diseases.


Subject(s)
Antibodies, Viral , Autoantibodies , COVID-19 , Cross Reactions , Epitopes , Molecular Mimicry , SARS-CoV-2 , Systemic Inflammatory Response Syndrome , Child , Humans , Antibodies, Viral/immunology , Autoantibodies/immunology , Coronavirus Nucleocapsid Proteins/chemistry , Coronavirus Nucleocapsid Proteins/immunology , COVID-19/immunology , COVID-19/virology , COVID-19/complications , Cross Reactions/immunology , Epitopes/immunology , Epitopes/chemistry , Molecular Mimicry/immunology , Phosphoproteins/chemistry , Phosphoproteins/immunology , SARS-CoV-2/chemistry , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Sorting Nexins/chemistry , Sorting Nexins/immunology , Systemic Inflammatory Response Syndrome/immunology , Systemic Inflammatory Response Syndrome/pathology , Systemic Inflammatory Response Syndrome/virology , T-Lymphocytes/immunology
16.
Yi Chuan ; 46(8): 649-660, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39140145

ABSTRACT

The localization of the meiotic specific regulatory molecule Moa1 to the centromere is regulated by the kinetochore protein CENP-C, and participates in the cohesion of sister chromatids in the centromere region mediated by the cohesin Rec8. To examine the interaction of these proteins, we analyzed the interactions between Moa1 and Rec8, CENP-C by yeast two-hybrid assays and identified several amino acid residues in Moa1 required for the interaction with CENP-C and Rec8. The results revealed that the interaction between Moa1 and CENP-C is crucial for the Moa1 to participate in the regulation of monopolar attachment of sister kinetochores. However, mutation at S143 and T150 of Moa1, which are required for interaction with Rec8 in the two-hybrid assay, did not show significant defects. Mutations in amino acid residues may not be sufficient to interfere with the interaction between Moa1 and Rec8 in vivo. Further research is needed to determine the interaction domain between Moa1 and Rec8. This study revealed specific amino acid sites at which Moa1 affects the meiotic homologous chromosome segregation, providing a deeper understanding of the mechanism of meiotic chromosome segregation.


Subject(s)
Chromosomal Proteins, Non-Histone , Meiosis , Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Protein Binding , Kinetochores/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Two-Hybrid System Techniques , Chromosome Segregation , Cohesins , Phosphoproteins
17.
Cell Rep Methods ; 4(8): 100839, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39127042

ABSTRACT

The availability of data from profiling of cancer patients with multiomics is rapidly increasing. However, integrative analysis of such data for personalized target identification is not trivial. Multiomics2Targets is a platform that enables users to upload transcriptomics, proteomics, and phosphoproteomics data matrices collected from the same cohort of cancer patients. After uploading the data, Multiomics2Targets produces a report that resembles a research publication. The uploaded matrices are processed, analyzed, and visualized using the tools Enrichr, KEA3, ChEA3, Expression2Kinases, and TargetRanger to identify and prioritize proteins, genes, and transcripts as potential targets. Figures and tables, as well as descriptions of the methods and results, are automatically generated. Reports include an abstract, introduction, methods, results, discussion, conclusions, and references and are exportable as citable PDFs and Jupyter Notebooks. Multiomics2Targets is applied to analyze version 3 of the Clinical Proteomic Tumor Analysis Consortium (CPTAC3) pan-cancer cohort, identifying potential targets for each CPTAC3 cancer subtype. Multiomics2Targets is available from https://multiomics2targets.maayanlab.cloud/.


Subject(s)
Neoplasms , Phosphoproteins , Proteomics , Transcriptome , Humans , Proteomics/methods , Neoplasms/genetics , Neoplasms/metabolism , Phosphoproteins/metabolism , Phosphoproteins/genetics , Cohort Studies , Gene Expression Profiling/methods , Software , Computational Biology/methods
18.
Methods Mol Biol ; 2823: 11-25, 2024.
Article in English | MEDLINE | ID: mdl-39052211

ABSTRACT

The sensitivity of phosphorylation site identification by mass spectrometry (MS)-based phosphoproteomics has improved significantly. However, the lack of kinase-substrate relationship (KSR) data has hindered improvement of the range and accuracy of kinase activity prediction using phosphoproteome data. We herein describe the application of a systematic identification of KSR by integrated phosphoproteome and interactome analysis using doxycycline (Dox)-induced target kinase-overexpressing HEK-293 cells.


Subject(s)
Phosphoproteins , Proteome , Proteomics , Humans , Phosphoproteins/metabolism , Phosphoproteins/analysis , HEK293 Cells , Proteomics/methods , Phosphorylation , Proteome/metabolism , Substrate Specificity , Mass Spectrometry/methods , Protein Kinases/metabolism , Protein Interaction Mapping/methods , Doxycycline/pharmacology
19.
J Med Virol ; 96(8): e29826, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39056254

ABSTRACT

Commercially available assays for measuring severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) anti-spike (S) or anti-nucleocapsid (N) antibodies differ in units, making results comparisons challenging. This study aimed to develop conversion equations between five quantitative anti-S antibody tests and to assess the agreement over time between three qualitative anti-N antibody tests. Blood samples from 24 216 vaccinated healthcare workers in Hiroshima Prefecture, Japan, were analyzed for anti-S antibodies using five quantitative tests (Abbott, Fujirebio, Ortho, Sysmex, Roche) and for anti-N antibodies using three qualitative tests (Abbott, Sysmex, Roche). Geometric mean regression was performed to establish equations for converting measured values between the five quantitative tests. Fleiss κ statistic was used to assess the agreement between the three qualitative tests. A strong correlation (Pearson's coefficient r > 0.9) was found for each pair of the five quantitative tests measuring anti-S antibodies, enabling the development of equations to convert values between each pair. Using these equations, which are based on the original output unit of each test, values obtained from one test can be transformed to be equivalent to the corresponding values in another test. For the three tests for anti-N antibodies, the agreement was substantial in the total sample (Fleiss' κ, 0.74) and moderate among those with self-reported past coronavirus disease 2019 (COVID-19) infection (Fleiss' κ, 0.39). The agreement decreased with time after infection. Reduced agreement between anti-N antibodies tests over time suggests caution in comparing seroepidemiological studies of COVID-19 exposure based on anti-N antibodies measurement. The findings could help improve antibody measurement systems and inform public health decision-makers.


Subject(s)
Antibodies, Viral , COVID-19 Serological Testing , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , Spike Glycoprotein, Coronavirus/immunology , Antibodies, Viral/blood , COVID-19/diagnosis , COVID-19/immunology , SARS-CoV-2/immunology , COVID-19 Serological Testing/methods , Japan , Coronavirus Nucleocapsid Proteins/immunology , Health Personnel , Phosphoproteins
20.
Viruses ; 16(7)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39066279

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of COVID-19 and responsible for the global coronavirus pandemic which started in 2019. Despite exhaustive efforts to trace its origins, including potential links with pangolins and bats, the precise origins of the virus remain unclear. Bats have been recognized as natural hosts for various coronaviruses, including the Middle East respiratory coronavirus (MERS-CoV) and the SARS-CoV. This study presents a comparative analysis of the SARS-CoV-2 nucleocapsid protein (N) interactome in human and bat cell lines. We identified approximately 168 cellular proteins as interacting partners of SARS-CoV-2 N in human cells and 196 cellular proteins as interacting partners with this protein in bat cells. The results highlight pathways and events that are both common and unique to either bat or human cells. Understanding these interactions is crucial to comprehend the reasons behind the remarkable resilience of bats to viral infections. This study provides a foundation for a deeper understanding of host-virus interactions in different reservoirs.


Subject(s)
COVID-19 , Chiroptera , Coronavirus Nucleocapsid Proteins , Phosphoproteins , Proteomics , SARS-CoV-2 , Chiroptera/virology , Humans , SARS-CoV-2/metabolism , Animals , Coronavirus Nucleocapsid Proteins/metabolism , Cell Line , Proteomics/methods , Phosphoproteins/metabolism , COVID-19/virology , COVID-19/metabolism , Host-Pathogen Interactions , Protein Interaction Maps
SELECTION OF CITATIONS
SEARCH DETAIL