Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 55
Filtrer
1.
Commun Biol ; 7(1): 704, 2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38851817

RÉSUMÉ

Aspergillus fumigatus represents a public health problem due to the high mortality rate in immunosuppressed patients and the emergence of antifungal-resistant isolates. Protein acetylation is a crucial post-translational modification that controls gene expression and biological processes. The strategic manipulation of enzymes involved in protein acetylation has emerged as a promising therapeutic approach for addressing fungal infections. Sirtuins, NAD+-dependent lysine deacetylases, regulate protein acetylation and gene expression in eukaryotes. However, their role in the human pathogenic fungus A. fumigatus remains unclear. This study constructs six single knockout strains of A. fumigatus and a strain lacking all predicted sirtuins (SIRTKO). The mutant strains are viable under laboratory conditions, indicating that sirtuins are not essential genes. Phenotypic assays suggest sirtuins' involvement in cell wall integrity, secondary metabolite production, thermotolerance, and virulence. Deletion of sirE attenuates virulence in murine and Galleria mellonella infection models. The absence of SirE alters the acetylation status of proteins, including histones and non-histones, and triggers significant changes in the expression of genes associated with secondary metabolism, cell wall biosynthesis, and virulence factors. These findings encourage testing sirtuin inhibitors as potential therapeutic strategies to combat A. fumigatus infections or in combination therapy with available antifungals.


Sujet(s)
Aspergillose , Aspergillus fumigatus , Sirtuines , Aspergillus fumigatus/pathogénicité , Aspergillus fumigatus/génétique , Aspergillus fumigatus/enzymologie , Sirtuines/génétique , Sirtuines/métabolisme , Virulence , Animaux , Souris , Aspergillose/microbiologie , Aspergillose/traitement médicamenteux , Acétylation , Protéines fongiques/génétique , Protéines fongiques/métabolisme , Régulation de l'expression des gènes fongiques , Facteurs de virulence/génétique , Facteurs de virulence/métabolisme , Papillons de nuit/microbiologie
2.
J Proteome Res ; 23(6): 2148-2159, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38785273

RÉSUMÉ

Diverse proteomics-based strategies have been applied to saliva to quantitatively identify diagnostic and prognostic targets for oral cancer. Considering that these targets may be regulated by events that do not imply variation in protein abundance levels, we hypothesized that changes in protein conformation can be associated with diagnosis and prognosis, revealing biological processes and novel targets of clinical relevance. For this, we employed limited proteolysis-mass spectrometry in saliva samples to explore structural alterations, comparing the proteome of healthy control and oral squamous cell carcinoma (OSCC) patients with and without lymph node metastasis. Thirty-six proteins with potential structural rearrangements were associated with clinical patient features including transketolase and its interacting partners. Moreover, N-glycosylated peptides contribute to structural rearrangements of potential diagnostic and prognostic markers. Altogether, this approach utilizes saliva proteins to search for targets for diagnosing and prognosing oral cancer and can guide the discovery of potential regulated sites beyond protein-level abundance.


Sujet(s)
Tumeurs de la bouche , Protéome , Salive , Humains , Tumeurs de la bouche/métabolisme , Tumeurs de la bouche/anatomopathologie , Tumeurs de la bouche/diagnostic , Salive/composition chimique , Salive/métabolisme , Protéome/analyse , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/diagnostic , Femelle , Marqueurs biologiques tumoraux/métabolisme , Mâle , Métastase lymphatique , Conformation des protéines , Adulte d'âge moyen , Pronostic , Protéomique/méthodes , Transketolase/métabolisme , Sujet âgé , Spectrométrie de masse , Protéines et peptides salivaires/métabolisme , Protéines et peptides salivaires/analyse
3.
Cytotherapy ; 26(5): 444-455, 2024 05.
Article de Anglais | MEDLINE | ID: mdl-38363248

RÉSUMÉ

BACKGROUND AIMS: Coronavirus disease 2019 (COVID-19) is characterized by a broad spectrum of clinical manifestations with the potential to progress to multiple organ dysfunction in severe cases. Extracellular vesicles (EVs) carry a range of biological cargoes, which may be used as biomarkers of disease state. METHODS: An exploratory secondary analysis of the SARITA-2 and SARITA-1 datasets (randomized clinical trials on patients with mild and moderate/severe COVID-19) was performed. Serum-derived EVs were used for proteomic analysis to identify enriched biological processes and key proteins, thus providing insights into differences in disease severity. Serum-derived EVs were separated from patients with COVID-19 by size exclusion chromatography and nanoparticle tracking analysis was used to determine particle concentration and diameter. Liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) was performed to identify and quantify protein signatures. Bioinformatics and multivariate statistical analysis were applied to distinguish candidate proteins associated with disease severity (mild versus moderate/severe COVID-19). RESULTS: No differences were observed in terms of the concentration and diameter of enriched EVs between mild (n = 14) and moderate/severe (n = 30) COVID-19. A total of 414 proteins were found to be present in EVs, of which 360 were shared while 48 were uniquely present in severe/moderate compared to mild COVID-19. The main biological signatures in moderate/severe COVID-19 were associated with platelet degranulation, exocytosis, complement activation, immune effector activation, and humoral immune response. Von Willebrand factor, serum amyloid A-2 protein, histone H4 and H2A type 2-C, and fibrinogen ß-chain were the most differentially expressed proteins between severity groups. CONCLUSION: Exploratory proteomic analysis of serum-derived EVs from patients with COVID-19 detected key proteins related to immune response and activation of coagulation and complement pathways, which are associated with disease severity. Our data suggest that EV proteins may be relevant biomarkers of disease state and prognosis.


Sujet(s)
COVID-19 , Vésicules extracellulaires , Protéomique , SARS-CoV-2 , Indice de gravité de la maladie , Humains , COVID-19/sang , COVID-19/diagnostic , COVID-19/immunologie , Vésicules extracellulaires/métabolisme , Protéomique/méthodes , Femelle , Mâle , Adulte d'âge moyen , Marqueurs biologiques/sang , Sujet âgé , Adulte , Spectrométrie de masse en tandem , Chromatographie en phase liquide
4.
Mol Cell Proteomics ; 22(12): 100676, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37940003

RÉSUMÉ

Extracellular vesicles (EVs) are biomolecule carriers for intercellular communication in health and disease. Nef is a HIV virulence factor that is released from cells within EVs and is present in plasma EVs of HIV-1 infected individuals. We performed a quantitative proteomic analysis to fully characterize the Nef-induced changes in protein composition of T cell-derived EVs and identify novel host targets of HIV. Several proteins with well-described roles in infection or not previously associated with HIV pathogenesis were specifically modulated by Nef in EVs. Among the downregulated proteins are the interferon-induced transmembrane 1, 2, and 3 (IFITM1-3) proteins, broad-spectrum antiviral factors known to be cell-to-cell transferable by EVs. We demonstrate that Nef depletes IFITM1-3 from EVs by excluding these proteins from the plasma membrane and lipid rafts, which are sites of EVs biogenesis in T cells. Our data establish Nef as a modulator of EVs' global protein content and as an HIV factor that antagonizes IFITMs.


Sujet(s)
Vésicules extracellulaires , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Humains , Lymphocytes T , Protéome/métabolisme , Protéomique , Vésicules extracellulaires/métabolisme , Interférons/métabolisme , Infections à VIH/métabolisme , Antiviraux/métabolisme
5.
Int J Mol Sci ; 24(9)2023 May 04.
Article de Anglais | MEDLINE | ID: mdl-37175936

RÉSUMÉ

The effects of the administration of mesenchymal stromal cells (MSC) may vary according to the source. We hypothesized that MSC-derived extracellular vesicles (EVs) obtained from bone marrow (BM), adipose (AD), or lung (L) tissues may also lead to different effects in sepsis. We profiled the proteome from EVs as a first step toward understanding their mechanisms of action. Polymicrobial sepsis was induced in C57BL/6 mice by cecal ligation and puncture (SEPSIS) and SHAM (control) animals only underwent laparotomy. Twenty-four hours after surgery, animals in the SEPSIS group were randomized to receive saline or 3 × 106 MSC-derived EVs from BM, AD, or L. The diffuse alveolar damage was decreased with EVs from all three sources. In kidneys, BM-, AD-, and L-EVs reduced edema and expression of interleukin-18. Kidney injury molecule-1 expression decreased only in BM- and L-EVs groups. In the liver, only BM-EVs reduced congestion and cell infiltration. The size and number of EVs from different sources were not different, but the proteome of the EVs differed. BM-EVs were enriched for anti-inflammatory proteins compared with AD-EVs and L-EVs. In conclusion, BM-EVs were associated with less organ damage compared with the other sources of EVs, which may be related to differences detected in their proteome.


Sujet(s)
Vésicules extracellulaires , Cellules souches mésenchymateuses , Sepsie , Animaux , Souris , Vésicules extracellulaires/métabolisme , Poumon , Cellules souches mésenchymateuses/métabolisme , Souris de lignée C57BL , Protéome/métabolisme , Sepsie/métabolisme
6.
Biochem Soc Trans ; 51(2): 771-781, 2023 04 26.
Article de Anglais | MEDLINE | ID: mdl-37021691

RÉSUMÉ

Cancer is a significant cause of death, precluding increasing life expectancy worldwide. That is a multifactorial disease initiated by intrinsic or extrinsic factors that induce cell differentiation into cancer cells. However, cancer development, progression, and metastasis are not controlled only by cancer cells. The entire environment around these cells, named tumor microenvironment (TME), influences tumor development and spread. The tumor microenvironment is formed by cancer cells and heterogenous nonmalignant cells integrated with a complex extracellular matrix. The main cellular components of the TME are cancer-associated fibroblasts (CAFs), T lymphocytes, B cells, tumor-associated macrophages (TAMs), dendritic cells (DC), natural killer (NK) cells, tumor-associated neutrophils (TANs), Stem Cells, Endothelial Cells and their soluble secreted extracellular vesicles (EVs) that modulate cancer cells to establish and disseminate. This review provides a recent insight into the role of EVs secreted from different populations of the TME associated with the initiation and progression of carcinoma.


Sujet(s)
Fibroblastes associés au cancer , Carcinomes , Vésicules extracellulaires , Humains , Microenvironnement tumoral , Cellules endothéliales , Lymphocytes B
7.
J Clin Periodontol ; 50(7): 942-951, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-36935103

RÉSUMÉ

AIM: To compare the salivary proteomic profile of periodontitis-affected (PA) parents and their offspring to periodontally healthy (PH) dyads in the pursuit of possible biomarkers for early diagnosis of this disease. MATERIALS AND METHODS: Unstimulated saliva samples collected from 17 pairs of PA or PH individuals and their children were submitted to mass spectrometric analyses followed by proteomic analyses. Primary PA fibroblasts were triggered towards having an inflammatory response, and an immunoenzymatic assay of its supernatant was performed to validate the obtained data. RESULTS: ANXA1, KRT4, GSTP1, HPX, A2M and KRT13 were lower in PA parents and their children, and IGHG1, CSTB, KRT9, SMR3B, IGHG4 and SERPINA1 were higher. ANXA1 presented the highest fold change, 7.1 times less produced in children of PA parents, and was selected as a potential biomarker for periodontitis. The in vitro assay also showed lower ANXA1 production by cells of PA patients. CONCLUSION: Before any clinical sign of periodontal loss, descendants of PA patients have an altered proteomic profile compared to PH individuals, presenting a lower abundance of ANXA1. This protein is suggested as a potential biomarker for periodontitis.


Sujet(s)
Annexine A1 , Parodontite , Enfant , Humains , Annexine A1/analyse , Annexine A1/métabolisme , Marqueurs biologiques/métabolisme , Parodontite/diagnostic , Parodontite/métabolisme , Protéomique , Salive/composition chimique
8.
Nat Commun ; 13(1): 6725, 2022 11 07.
Article de Anglais | MEDLINE | ID: mdl-36344512

RÉSUMÉ

The poor prognosis of head and neck cancer (HNC) is associated with metastasis within the lymph nodes (LNs). Herein, the proteome of 140 multisite samples from a 59-HNC patient cohort, including primary and matched LN-negative or -positive tissues, saliva, and blood cells, reveals insights into the biology and potential metastasis biomarkers that may assist in clinical decision-making. Protein profiles are strictly associated with immune modulation across datasets, and this provides the basis for investigating immune markers associated with metastasis. The proteome of LN metastatic cells recapitulates the proteome of the primary tumor sites. Conversely, the LN microenvironment proteome highlights the candidate prognostic markers. By integrating prioritized peptide, protein, and transcript levels with machine learning models, we identify nodal metastasis signatures in blood and saliva. We present a proteomic characterization wiring multiple sites in HNC, thus providing a promising basis for understanding tumoral biology and identifying metastasis-associated signatures.


Sujet(s)
Tumeurs de la tête et du cou , Protéome , Humains , Métastase lymphatique/anatomopathologie , Protéomique , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/anatomopathologie , Noeuds lymphatiques/anatomopathologie , Microenvironnement tumoral
10.
J Proteomics ; 254: 104474, 2022 03 15.
Article de Anglais | MEDLINE | ID: mdl-34990821

RÉSUMÉ

Syndecans belong to the family of transmembrane heparan sulfate proteoglycans and are associated with many physiopathological processes, including oral cancer. As previously shown soluble syndecan-1 (SDC1) fragments and synthetic SDC1 peptide were able to induce cell migration in oral cancer cell lines. In order to explore the role of SDC1 in oral cancer, we have investigated SDC1 interacting partners and its functional role in oral cancer models. Here we have shown that SDC1 interacts with follistatin-related protein 1 (FSTL1) by its ectodomain (ectoSDC1) and extracellular juxtamembrane peptide (pepSDC1) and that their transcript levels can affect tumor events. Using orthotopic mouse model we identified that the knock-down for FSTL1 (shFSTL1) or for both FSTL1 and SDC1 (sh2KD) produced less aggressive and infiltrative tumors, with lower keratinization deposition, but with increased levels of epithelial-mesenchymal transition and proliferation compared to control and SDC1 knock-down. Based on cell culture assays, we suggest that the shFSTL1 effect on tumor tissues might be from significant increase of mRNA levels of Activin A (ActA) and its resceptors. This study shows for the first time two different complexes, SDC1 and FSTL1; pepSDC1 and FSTL1, exhibiting a close relationship in cell signaling events, as FSTL1 promotes a more aggressive phenotype. SIGNIFICANCE: This work contributes to the understanding of new SDC1 functions, based on the investigation of protein-protein complex formation in Oral Squamous cell carcinoma (OSCC) models. The FSTL1 identification, as an interacting partner of SDC1 ectodomain and of its derived peptide promotes molecular events that favors cancer development and progression, as highlighted by Activin A (ActA) and Epithelial-mesenchymal transition (EMT) gene expression and by changes in the phenotype of orthotopic OSCC mouse tumor tissues when SDC1-FSTL1 expression is modulated.


Sujet(s)
Carcinome épidermoïde , Protéines apparentées à la follistatine , Tumeurs de la tête et du cou , Tumeurs de la bouche , Animaux , Protéines apparentées à la follistatine/génétique , Souris , Phénotype , Carcinome épidermoïde de la tête et du cou , Syndécane-1/génétique , Syndécane-1/métabolisme
11.
Nat Commun ; 12(1): 3038, 2021 05 24.
Article de Anglais | MEDLINE | ID: mdl-34031424

RÉSUMÉ

Mayaro virus (MAYV) is an emerging arbovirus of the Americas that may cause a debilitating arthritogenic disease. The biology of MAYV is not fully understood and largely inferred from related arthritogenic alphaviruses. Here, we present the structure of MAYV at 4.4 Å resolution, obtained from a preparation of mature, infective virions. MAYV presents typical alphavirus features and organization. Interactions between viral proteins that lead to particle formation are described together with a hydrophobic pocket formed between E1 and E2 spike proteins and conformational epitopes specific of MAYV. We also describe MAYV glycosylation residues in E1 and E2 that may affect MXRA8 host receptor binding, and a molecular "handshake" between MAYV spikes formed by N262 glycosylation in adjacent E2 proteins. The structure of MAYV is suggestive of structural and functional complexity among alphaviruses, which may be targeted for specificity or antiviral activity.


Sujet(s)
Infections à alphavirus/virologie , Alphavirus/ultrastructure , Cryomicroscopie électronique , Spectrométrie de masse , Alphavirus/immunologie , Infections à alphavirus/immunologie , Animaux , Anticorps neutralisants , Chlorocebus aethiops , Glycosylation , Humains , Immunoglobulines , Protéines membranaires , Cellules Vero
12.
Biochim Biophys Acta Proteins Proteom ; 1869(8): 140659, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-33839314

RÉSUMÉ

Saliva is a biofluid that maintains the health of oral tissues and the homeostasis of oral microbiota. Studies have demonstrated that Oral squamous cell carcinoma (OSCC) patients have different salivary microbiota than healthy individuals. However, the relationship between these microbial differences and clinicopathological outcomes is still far from conclusive. Herein, we investigate the capability of using metagenomic and metaproteomic saliva profiles to distinguish between Control (C), OSCC without active lesion (L0), and OSCC with active lesion (L1) patients. The results show that there are significantly distinct taxonomies and functional changes in L1 patients compared to C and L0 patients, suggesting compositional modulation of the oral microbiome, as the relative abundances of Centipeda, Veillonella, and Gemella suggested by metagenomics are correlated with tumor size, clinical stage, and active lesion. Metagenomics results also demonstrated that poor overall patient survival is associated with a higher relative abundance of Stenophotromonas, Staphylococcus, Centipeda, Selenomonas, Alloscordovia, and Acitenobacter. Finally, compositional and functional differences in the saliva content by metaproteomics analysis can distinguish healthy individuals from OSCC patients. In summary, our study suggests that oral microbiota and their protein abundance have potential diagnosis and prognosis value for oral cancer patients. Further studies are necessary to understand the role of uniquely detected metaproteins in the microbiota of healthy and OSCC patients as well as the crosstalk between saliva host proteins and the oral microbiome present in OSCC.


Sujet(s)
Salive/microbiologie , Carcinome épidermoïde de la tête et du cou/microbiologie , Adulte , Sujet âgé , Études de cohortes , Femelle , Humains , Mâle , Métagénomique/méthodes , Microbiote/génétique , Adulte d'âge moyen , Tumeurs de la bouche/métabolisme , Tumeurs de la bouche/microbiologie , Pronostic , Protéomique/méthodes , Carcinome épidermoïde de la tête et du cou/métabolisme
13.
Mol Cell Proteomics ; 20: 100004, 2021.
Article de Anglais | MEDLINE | ID: mdl-33578082

RÉSUMÉ

Protease activity has been associated with pathological processes that can lead to cancer development and progression. However, understanding the pathological unbalance in proteolysis is challenging because changes can occur simultaneously at protease, their inhibitor, and substrate levels. Here, we present a pipeline that combines peptidomics, proteomics, and peptidase predictions for studying proteolytic events in the saliva of 79 patients and their association with oral squamous cell carcinoma (OSCC) prognosis. Our findings revealed differences in the saliva peptidome of patients with (pN+) or without (pN0) lymph-node metastasis and delivered a panel of ten endogenous peptides correlated with poor prognostic factors plus five molecules able to classify pN0 and pN+ patients (area under the receiver operating characteristic curve > 0.85). In addition, endopeptidases and exopeptidases putatively implicated in the processing of differential peptides were investigated using cancer tissue gene expression data from public repositories, reinforcing their association with poorer survival rates and prognosis in oral cancer. The dynamics of the OSCC-related proteolysis were further explored via the proteomic profiling of saliva. This revealed that peptidase/endopeptidase inhibitors exhibited reduced levels in the saliva of pN+ patients, as confirmed by selected reaction monitoring-mass spectrometry, while minor changes were detected in the level of saliva proteases. Taken together, our results indicated that proteolytic activity is accentuated in the saliva of patients with OSCC and lymph-node metastasis and, at least in part, is modulated by reduced levels of salivary peptidase inhibitors. Therefore, this integrated pipeline provided better comprehension and discovery of molecular features with implications in the oral cancer metastasis prognosis.


Sujet(s)
Carcinome épidermoïde/métabolisme , Métastase lymphatique , Tumeurs de la bouche/métabolisme , Peptide hydrolases/métabolisme , Peptides/analyse , Salive/composition chimique , Carcinome épidermoïde/anatomopathologie , Humains , Tumeurs de la bouche/anatomopathologie , Peptides/métabolisme , Pronostic , Protéomique
14.
Sci Rep ; 11(1): 1995, 2021 01 21.
Article de Anglais | MEDLINE | ID: mdl-33479267

RÉSUMÉ

The joint disease called pararamosis is an occupational disease caused by accidental contact with bristles of the caterpillar Premolis semirufa. The chronic inflammatory process narrows the joint space and causes alterations in bone structure and cartilage degeneration, leading to joint stiffness. Aiming to determine the bristle components that could be responsible for this peculiar envenomation, in this work we have examined the toxin composition of the caterpillar bristles extract and compared it with the differentially expressed genes (DEGs) in synovial biopsies of patients affected with rheumatoid arthritis (RA) and osteoarthritis (OA). Among the proteins identified, 129 presented an average of 63% homology with human proteins and shared important conserved domains. Among the human homologous proteins, we identified seven DEGs upregulated in synovial biopsies from RA or OA patients using meta-analysis. This approach allowed us to suggest possible toxins from the pararama bristles that could be responsible for starting the joint disease observed in pararamosis. Moreover, the study of pararamosis, in turn, may lead to the discovery of specific pharmacological targets related to the early stages of articular diseases.


Sujet(s)
Polyarthrite rhumatoïde/épidémiologie , Maladies articulaires/épidémiologie , Lepidoptera/pathogénicité , Arthrose/épidémiologie , Toxines biologiques/toxicité , Animaux , Polyarthrite rhumatoïde/induit chimiquement , Humains , Inflammation/induit chimiquement , Inflammation/épidémiologie , Maladies articulaires/induit chimiquement , Maladies articulaires/anatomopathologie , Lepidoptera/composition chimique , Maladies professionnelles/induit chimiquement , Maladies professionnelles/épidémiologie , Arthrose/induit chimiquement , Membrane synoviale/effets des médicaments et des substances chimiques , Membrane synoviale/anatomopathologie , Toxines biologiques/isolement et purification , Venins/effets indésirables , Venins/composition chimique
15.
Oral Dis ; 27(4): 952-961, 2021 May.
Article de Anglais | MEDLINE | ID: mdl-32772410

RÉSUMÉ

OBJECTIVE: Odontogenic keratocyst (OKC) is a benign lesion that tends to recur after surgical treatment. In an attempt to clarify the molecular basis underlining the OKC pathobiology, we aimed to analyze its proteomic profile. MATERIALS AND METHODS: We compared the proteomic profiles of five OKC and matched normal oral mucosa by using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Then, we performed enrichment analysis and a literature search for the immunoexpression of the proteomics targets. RESULTS: We identified 1,150 proteins and 72 differently expressed proteins (log2 fold change ≥ 1.5; p < .05). Twenty-seven peptides were exclusively detected in the OKC samples. We found 35 enriched pathways related to cell differentiation and tissue architecture, including keratinocyte differentiation, keratinization, desmosome, and extracellular matrix (ECM) organization and degradation. The immunoexpression information of 11 out of 50 proteins identified in the enriched pathways was obtained. We found the downregulation of four desmosomal proteins (JUP, PKP1, PKP3, and PPL) and upregulation of ECM proteases (MMP-2, MMP-9, and cathepsins). CONCLUSIONS: Proteomic analysis strengthened the notion that OKC cells have a similar proteomic profile to oral keratinocytes. Contextual investigation of the differentially expressed proteins revealed the deregulation of desmosome proteins and ECM degradation as important alterations in OKC pathobiology.


Sujet(s)
Kystes odontogènes , Peptide hydrolases , Chromatographie en phase liquide , Matrice extracellulaire , Humains , Récidive tumorale locale , Protéomique , Spectrométrie de masse en tandem
16.
Sci Rep, v. 11, 1995, jan. 2021
Article de Anglais | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: bud-3484

RÉSUMÉ

The joint disease called pararamosis is an occupational disease caused by accidental contact with bristles of the caterpillar Premolis semirufa. The chronic inflammatory process narrows the joint space and causes alterations in bone structure and cartilage degeneration, leading to joint stiffness. Aiming to determine the bristle components that could be responsible for this peculiar envenomation, in this work we have examined the toxin composition of the caterpillar bristles extract and compared it with the differentially expressed genes (DEGs) in synovial biopsies of patients affected with rheumatoid arthritis (RA) and osteoarthritis (OA). Among the proteins identified, 129 presented an average of 63% homology with human proteins and shared important conserved domains. Among the human homologous proteins, we identified seven DEGs upregulated in synovial biopsies from RA or OA patients using meta-analysis. This approach allowed us to suggest possible toxins from the pararama bristles that could be responsible for starting the joint disease observed in pararamosis. Moreover, the study of pararamosis, in turn, may lead to the discovery of specific pharmacological targets related to the early stages of articular diseases.

17.
Redox Biol ; 37: 101735, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-33011677

RÉSUMÉ

The activity of Thioredoxin-1 (Trx-1) is adjusted by the balance of its monomeric, active and its dimeric, inactive state. The regulation of this balance is not completely understood. We have previously shown that the cytoplasmic domain of the transmembrane protein A Disintegrin And Metalloprotease 17 (ADAM17cyto) binds to Thioredoxin-1 (Trx-1) and the destabilization of this interaction favors the dimeric state of Trx-1. Here, we investigate whether ADAM17 plays a role in the conformation and activation of Trx-1. We found that disrupting the interacting interface with Trx-1 by a site-directed mutagenesis in ADAM17 (ADAM17cytoF730A) caused a decrease of Trx-1 reductive capacity and activity. Moreover, we observed that ADAM17 overexpressing cells favor the monomeric state of Trx-1 while knockdown cells do not. As a result, there is a decrease of cell oxidant levels and ADAM17 sheddase activity and an increase in the reduced cysteine-containing peptides in intracellular proteins in ADAM17cyto overexpressing cells. A mechanistic explanation that ADAM17cyto favors the monomeric, active state of Trx-1 is the formation of a disulfide bond between Cys824 at the C-terminal of ADAM17cyto with the Cys73 of Trx-1, which is involved in the dimerization site of Trx-1. In summary, we propose that ADAM17 is able to modulate Trx-1 conformation affecting its activity and intracellular redox state, bringing up a novel possibility for positive regulation of thiol isomerase activity in the cell by mammalian metalloproteinases.


Sujet(s)
Protéine ADAM17 , Cystéine , Thiorédoxines , Cystéine/métabolisme , Cellules HEK293 , Humains , Conformation moléculaire , Oxydoréduction , Thiols , Thiorédoxines/génétique , Thiorédoxines/métabolisme
18.
Oral Dis ; 26(8): 1610-1618, 2020 Nov.
Article de Anglais | MEDLINE | ID: mdl-31829479

RÉSUMÉ

Salivary gland tumors (SGTs) comprise a heterogeneous group of benign and malignant neoplasms that exhibit significant variability in their microscopic appearance, clinical presentation, and biological behavior. The etiologic factors are unknown; however, chromosomic translocation, secondary radiation, and chemotherapy can be associated with the development of SGT. It has been indicated that epigenetic alterations can be responsible for the development and progress of these neoplasms. The epigenetic mechanisms are defined as a set of DNA changes that do not alter the sequence of nucleotide bases but alter the expression of the proteins. These alterations have been studied in the SGT, and they were associated with the development and progress of these neoplasms and may influence on SGT prognosis. Hence, we critically review the currently available data on the participation of epigenetic events on salivary gland tumors.


Sujet(s)
Tumeurs des glandes salivaires , ADN , Épigenèse génétique , Humains , Pronostic , Tumeurs des glandes salivaires/génétique
19.
Redox Biol ; 18: 181-190, 2018 09.
Article de Anglais | MEDLINE | ID: mdl-30029165

RÉSUMÉ

Increased reactive oxygen species (ROS) formation may enhance matrix metalloproteinase (MMP)-2 activity and promote cardiovascular dysfunction. We show for the first time that MMP-2 is upstream of increased ROS formation and activates signaling mechanisms impairing redox balance. Incubation of vascular smooth muscle cells (VSMC) with recombinant MMP-2 increased ROS formation assessed with dihydroethidium (DHE) by flow cytometry. This effect was blocked by the antioxidant apocynin or by polyethylene glycol-catalase (PEG-catalase), and by MMP inhibitors (doxycycline or GM6001). Next, we showed in HEK293 cells that MMP-2 transactivates heparin-binding epidermal growth factor (HB-EGF) leading to EGF receptor (EGFR) activation and increased ROS concentrations. This effect was prevented by the EGFR kinase inhibitor Ag1478, and by phospholipase C (PLC) or protein kinase C (PKC) inhibitors (A778 or chelerythrine, respectively), confirming the involvement of EGFR pathway in MMP-2-induce responses. Next, we showed that intraluminal exposure of aortas to MMP-2 increased vascular MMP-2 levels detected by immunofluorescence and gelatinolytic activity (by in situ zimography) in association with increased ROS formation. This effect was inhibited by MMP inhibitors (phenanthroline or doxycycline) and by apocynin or PEG-catalase. MMP-2 also increased aortic contractility to phenylephrine and this effect was prevented by MMP inhibitor GM6001 and by apocynin or PEG-catalase, showing again that increased ROS formation mediates functional effects of MMP-2. These results show that MMP-2 activates the EGFR and triggers downstream signaling pathways increasing ROS formation and promoting vasoconstriction. These findings may have various implications for cardiovascular diseases.


Sujet(s)
Aorte/physiologie , Récepteurs ErbB/génétique , Matrix metalloproteinase 2/métabolisme , Muscles lisses vasculaires/physiologie , Activation de la transcription , Vasoconstriction , Animaux , Aorte/cytologie , Lignée cellulaire , Récepteurs ErbB/métabolisme , Mâle , Muscles lisses vasculaires/cytologie , Oxydoréduction , Lapins , Rats , Espèces réactives de l'oxygène/métabolisme
20.
Antioxid Redox Signal ; 29(8): 717-734, 2018 09 10.
Article de Anglais | MEDLINE | ID: mdl-29334756

RÉSUMÉ

AIMS: A disintegrin and metalloprotease 17 (ADAM17) modulates signaling events by releasing surface protein ectodomains such as TNFa and the EGFR-ligands. We have previously characterized cytoplasmic thioredoxin-1 (Trx-1) as a partner of ADAM17 cytoplasmic domain. Still, the mechanism of ADAM17 regulation by Trx-1 is unknown, and it has become of paramount importance to assess the degree of influence that Trx-1 has on metalloproteinase ADAM17. RESULTS: Combining discovery and targeted proteomic approaches, we uncovered that Trx-1 negatively regulates ADAM17 by direct and indirect effect. We performed cell-based assays with synthetic peptides and site-directed mutagenesis, and we demonstrated that the interaction interface of Trx-1 and ADAM17 is important for the negative regulation of ADAM17 activity. However, both Trx-1K72A and catalytic site mutant Trx-1C32/35S rescued ADAM17 activity, although the interaction with Trx-1C32/35S was unaffected, suggesting an indirect effect of Trx-1. We confirmed that the Trx-1C32/35S mutant showed diminished reductive capacity, explaining this indirect effect on increasing ADAM17 activity through oxidant levels. Interestingly, Trx-1K72A mutant showed similar oxidant levels to Trx-1C32/35S, even though its catalytic site was preserved. We further demonstrated that the general reactive oxygen species inhibitor, Nacetylcysteine (NAC), maintained the regulation of ADAM17 dependent of Trx-1 reductase activity levels; whereas the electron transport chain modulator, rotenone, abolished Trx-1 effect on ADAM17 activity. INNOVATION: We show for the first time that the mechanism of ADAM17 regulation, Trx-1 dependent, can be by direct interaction and indirect effect, bringing new insights into the cross-talk between isomerases and mammalian metalloproteinases. CONCLUSION: This unexpected Trx-1K72A behavior was due to more dimer formation and, consequently, the reduction of its Trx-1 reductase activity, evaluated through dimer verification, by gel filtration and mass spectrometry analysis. Antioxid. Redox Signal. 29, 717-734.


Sujet(s)
Protéine ADAM17/métabolisme , Thiorédoxines/métabolisme , Sites de fixation , Cellules HEK293 , Humains , Modèles moléculaires , Oxydoréduction , Thiorédoxines/analyse , Thiorédoxines/génétique , Cellules cancéreuses en culture
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE