Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 53
Filtrer
1.
Adv Sci (Weinh) ; 11(29): e2306860, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38864559

RÉSUMÉ

Breast tumor-initiating cells (BTICs) of triple-negative breast cancer (TNBC) tissues actively repair DNA and are resistant to treatments including chemotherapy, radiotherapy, and targeted therapy. Herein, it is found that a previously reported secreted protein, sclerostin domain containing 1 (SOSTDC1), is abundantly expressed in BTICs of TNBC cells and positively correlated with a poor patient prognosis. SOSTDC1 knockdown impairs homologous recombination (HR) repair, BTIC maintenance, and sensitized bulk cells and BTICs to Olaparib. Mechanistically, following Olaparib treatment, SOSTDC1 translocates to the nucleus in an importin-α dependent manner. Nuclear SOSTDC1 interacts with the N-terminus of the nucleoprotein, chromatin helicase DNA-binding factor (CHD1), to promote HR repair and BTIC maintenance. Furthermore, nuclear SOSTDC1 bound to ß-transducin repeat-containing protein (ß-TrCP) binding motifs of CHD1 is found, thereby blocking the ß-TrCP-CHD1 interaction and inhibiting ß-TrCP-mediated CHD1 ubiquitination and degradation. Collectively, these findings identify a novel nuclear SOSTDC1 pathway in regulating HR repair and BTIC maintenance, providing insight into the TNBC therapeutic strategies.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Protéines de liaison à l'ADN , Phtalazines , Pipérazines , Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/traitement médicamenteux , Femelle , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Pipérazines/pharmacologie , Phtalazines/pharmacologie , Souris , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Lignée cellulaire tumorale , Animaux , Résistance aux médicaments antinéoplasiques/génétique , Réparation de l'ADN par recombinaison/génétique , Évolution de la maladie , Cellules souches tumorales/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Noyau de la cellule/métabolisme , Helicase
2.
Nat Genet ; 56(7): 1377-1385, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38886586

RÉSUMÉ

The presence of basal lineage characteristics signifies hyperaggressive human adenocarcinomas of the breast, bladder and pancreas. However, the biochemical mechanisms that maintain this aberrant cell state are poorly understood. Here we performed marker-based genetic screens in search of factors needed to maintain basal identity in pancreatic ductal adenocarcinoma (PDAC). This approach revealed MED12 as a powerful regulator of the basal cell state in this disease. Using biochemical reconstitution and epigenomics, we show that MED12 carries out this function by bridging the transcription factor ΔNp63, a known master regulator of the basal lineage, with the Mediator complex to activate lineage-specific enhancer elements. Consistent with this finding, the growth of basal-like PDAC is hypersensitive to MED12 loss when compared to PDAC cells lacking basal characteristics. Taken together, our genetic screens have revealed a biochemical interaction that sustains basal identity in human cancer, which could serve as a target for tumor lineage-directed therapeutics.


Sujet(s)
Carcinome du canal pancréatique , Complexe médiateur , Tumeurs du pancréas , Facteurs de transcription , Protéines suppresseurs de tumeurs , Humains , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Complexe médiateur/génétique , Complexe médiateur/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme , Régulation de l'expression des gènes tumoraux , Lignée cellulaire tumorale , Lignage cellulaire/génétique , Éléments activateurs (génétique)
3.
Int J Biol Sci ; 20(6): 2130-2148, 2024.
Article de Anglais | MEDLINE | ID: mdl-38617541

RÉSUMÉ

Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer with limited effective therapeutic options readily available. We have previously demonstrated that lovastatin, an FDA-approved lipid-lowering drug, selectively inhibits the stemness properties of TNBC. However, the intracellular targets of lovastatin in TNBC remain largely unknown. Here, we unexpectedly uncovered ribosome biogenesis as the predominant pathway targeted by lovastatin in TNBC. Lovastatin induced the translocation of ribosome biogenesis-related proteins including nucleophosmin (NPM), nucleolar and coiled-body phosphoprotein 1 (NOLC1), and the ribosomal protein RPL3. Lovastatin also suppressed the transcript levels of rRNAs and increased the nuclear protein level and transcriptional activity of p53, a master mediator of nucleolar stress. A prognostic model generated from 10 ribosome biogenesis-related genes showed outstanding performance in predicting the survival of TNBC patients. Mitochondrial ribosomal protein S27 (MRPS27), the top-ranked risky model gene, was highly expressed and correlated with tumor stage and lymph node involvement in TNBC. Mechanistically, MRPS27 knockdown inhibited the stemness properties and the malignant phenotypes of TNBC. Overexpression of MRPS27 attenuated the stemness-inhibitory effect of lovastatin in TNBC cells. Our findings reveal that dysregulated ribosome biogenesis is a targetable vulnerability and targeting MRPS27 could be a novel therapeutic strategy for TNBC patients.


Sujet(s)
Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/génétique , Lovastatine/pharmacologie , Lovastatine/usage thérapeutique , Protéines ribosomiques/génétique , Protéines nucléaires , Ribosomes/génétique , Protéines mitochondriales
4.
Nat Commun ; 15(1): 3282, 2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38627380

RÉSUMÉ

Exposure to pathogens throughout a lifetime influences immunity and organ function. Here, we explore how the systemic host-response to bacterial urinary tract infection (UTI) induces tissue-specific alterations to the mammary gland. Utilizing a combination of histological tissue analysis, single cell transcriptomics, and flow cytometry, we identify that mammary tissue from UTI-bearing mice displays collagen deposition, enlarged ductal structures, ductal hyperplasia with atypical epithelial transcriptomes and altered immune composition. Bacterial cells are absent in the mammary tissue and blood of UTI-bearing mice, therefore, alterations to the distal mammary tissue are mediated by the systemic host response to local infection. Furthermore, broad spectrum antibiotic treatment resolves the infection and restores mammary cellular and tissue homeostasis. Systemically, unresolved UTI correlates with increased plasma levels of the metalloproteinase inhibitor, TIMP1, which controls extracellular matrix remodeling and neutrophil function. Treatment of nulliparous and post-lactation UTI-bearing female mice with a TIMP1 neutralizing antibody, restores mammary tissue normal homeostasis, thus providing evidence for a link between the systemic host response during UTI and mammary gland alterations.


Sujet(s)
Glandes mammaires animales , Infections urinaires , Animaux , Femelle , Souris , Collagène , Matrice extracellulaire/physiologie , Homéostasie
5.
Cancer Res ; 84(14): 2282-2296, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38657120

RÉSUMÉ

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with limited therapeutic options. IL1 receptor type 2 (IL1R2) promotes breast tumor-initiating cell (BTIC) self-renewal and tumor growth in TNBC, indicating that targeting it could improve patient treatment. In this study, we observed that IL1R2 blockade strongly attenuated macrophage recruitment and the polarization of tumor-associated macrophages (TAM) to inhibit BTIC self-renewal and CD8+ T-cell exhaustion, which resulted in reduced tumor burden and prolonged survival in TNBC mouse models. IL1R2 activation by TAM-derived IL1ß increased PD-L1 expression by interacting with the transcription factor Yin Yang 1 (YY1) and inducing YY1 ubiquitination and proteasomal degradation in both TAMs and TNBC cells. Loss of YY1 alleviated the transcriptional repression of c-Fos, which is a transcriptional activator of PDL-1. Combined treatment with an IL1R2-neutralizing antibodies and anti-PD-1 led to enhanced antitumor efficacy and reduced TAMs, BTICs, and exhausted CD8+ T cells. These results suggest that IL1R2 blockade might be a strategy to potentiate immune checkpoint blockade efficacy in TNBC to improve patient outcomes. Significance: IL1R2 in both macrophages and breast cancer cells orchestrates an immunosuppressive tumor microenvironment by upregulating PD-L1 expression and can be targeted to enhance the efficacy of anti-PD-1 in triple-negative breast cancer.


Sujet(s)
Tumeurs du sein triple-négatives , Tumeurs du sein triple-négatives/immunologie , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Animaux , Souris , Humains , Femelle , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Lymphocytes T CD8+/immunologie , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Macrophages associés aux tumeurs/immunologie , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/métabolisme , Facteur de transcription YY1/métabolisme , Facteur de transcription YY1/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Cellules souches tumorales/métabolisme , Cellules souches tumorales/immunologie , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/effets des médicaments et des substances chimiques
6.
Protein Cell ; 15(6): 419-440, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38437016

RÉSUMÉ

Tumor-resident microbiota in breast cancer promotes cancer initiation and malignant progression. However, targeting microbiota to improve the effects of breast cancer therapy has not been investigated in detail. Here, we evaluated the microbiota composition of breast tumors and found that enterotoxigenic Bacteroides fragilis (ETBF) was highly enriched in the tumors of patients who did not respond to taxane-based neoadjuvant chemotherapy. ETBF, albeit at low biomass, secreted the toxic protein BFT-1 to promote breast cancer cell stemness and chemoresistance. Mechanistic studies showed that BFT-1 directly bound to NOD1 and stabilized NOD1 protein. NOD1 was highly expressed on ALDH+ breast cancer stem cells (BCSCs) and cooperated with GAK to phosphorylate NUMB and promote its lysosomal degradation, thereby activating the NOTCH1-HEY1 signaling pathway to increase BCSCs. NOD1 inhibition and ETBF clearance increase the chemosensitivity of breast cancer by impairing BCSCs.


Sujet(s)
Bacteroides fragilis , Tumeurs du sein , Résistance aux médicaments antinéoplasiques , Cellules souches tumorales , Protéine adaptatrice de signalisation NOD1 , Humains , Protéine adaptatrice de signalisation NOD1/métabolisme , Protéine adaptatrice de signalisation NOD1/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/microbiologie , Tumeurs du sein/génétique , Femelle , Bacteroides fragilis/métabolisme , Bacteroides fragilis/génétique , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Toxines bactériennes/métabolisme , Toxines bactériennes/génétique , Animaux , Souris , Lignée cellulaire tumorale , Metalloendopeptidases
7.
Cancer Cell ; 42(3): 474-486.e12, 2024 Mar 11.
Article de Anglais | MEDLINE | ID: mdl-38402610

RÉSUMÉ

Chronic stress is associated with increased risk of metastasis and poor survival in cancer patients, yet the reasons are unclear. We show that chronic stress increases lung metastasis from disseminated cancer cells 2- to 4-fold in mice. Chronic stress significantly alters the lung microenvironment, with fibronectin accumulation, reduced T cell infiltration, and increased neutrophil infiltration. Depleting neutrophils abolishes stress-induced metastasis. Chronic stress shifts normal circadian rhythm of neutrophils and causes increased neutrophil extracellular trap (NET) formation via glucocorticoid release. In mice with neutrophil-specific glucocorticoid receptor deletion, chronic stress fails to increase NETs and metastasis. Furthermore, digesting NETs with DNase I prevents chronic stress-induced metastasis. Together, our data show that glucocorticoids released during chronic stress cause NET formation and establish a metastasis-promoting microenvironment. Therefore, NETs could be targets for preventing metastatic recurrence in cancer patients, many of whom will experience chronic stress due to their disease.


Sujet(s)
Pièges extracellulaires , Tumeurs du poumon , Humains , Animaux , Souris , Granulocytes neutrophiles/anatomopathologie , Tumeurs du poumon/anatomopathologie , Poumon/anatomopathologie , Microenvironnement tumoral
8.
Food Chem ; 444: 138624, 2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-38354655

RÉSUMÉ

A novel ratiometric fluorescent probe based on non-conjugated polymer dots (NCPDs) and gold nanocluster (AuNCs) was constructed to determine tert-butylhydroquinone (TBHQ). The probe exhibited dual emission peaks at 480 nm and 630 nm under 370 nm excitation. The fluorescence of AuNCs was quenched by TBHQ due to strong electrostatic interactions, whereas the emission of NCPDs increased. The ratio of fluorescence intensity at 480 nm to 630 nm (F480 / F630) was monitored as analytical signal response. The probe have been utilized for the detection of TBHQ with good linear relationship in the range of 0.2 to 60 µg/mL. The limit of detection (LOD) and the limit of quantitation (LOQ) were 0.048 µg/mL and 0.159 µg/L, respectively. Three levels of spiked-in TBHQ concentrations were obtained with recovery rates from 80 % to 102 %. The present study provided an effective ratiometric fluorescence method for selective screening of TBHQ in food samples.


Sujet(s)
Hydroquinones , Nanoparticules métalliques , Boîtes quantiques , Spectrométrie de fluorescence/méthodes , Polymères , Or , Colorants fluorescents
9.
Acta Pharm Sin B ; 13(4): 1686-1698, 2023 Apr.
Article de Anglais | MEDLINE | ID: mdl-37139408

RÉSUMÉ

Triple-negative breast cancer (TNBC) is a nasty disease with extremely high malignancy and poor prognosis. Annexin A3 (ANXA3) is a potential prognosis biomarker, displaying an excellent correlation of ANXA3 overexpression with patients' poor prognosis. Silencing the expression of ANXA3 effectively inhibits the proliferation and metastasis of TNBC, suggesting that ANXA3 can be a promising therapeutic target to treat TNBC. Herein, we report a first-in-class ANXA3-targeted small molecule (R)-SL18, which demonstrated excellent anti-proliferative and anti-invasive activities to TNBC cells. (R)-SL18 directly bound to ANXA3 and increased its ubiquitination, thereby inducing ANXA3 degradation with moderate family selectivity. Importantly, (R)-SL18 showed a safe and effective therapeutic potency in a high ANXA3-expressing TNBC patient-derived xenograft model. Furthermore, (R)-SL18 could reduce the ß-catenin level, and accordingly inhibit the Wnt/ß-catenin signaling pathway in TNBC cells. Collectively, our data suggested that targeting degradation of ANXA3 by (R)-SL18 possesses the potential to treat TNBC.

10.
Signal Transduct Target Ther ; 8(1): 97, 2023 03 01.
Article de Anglais | MEDLINE | ID: mdl-36859354

RÉSUMÉ

Our previous studies have showed that C-C motif chemokine ligand 20 (CCL20) advanced tumor progression and enhanced the chemoresistance of cancer cells by positively regulating breast cancer stem cell (BCSC) self-renewal. However, it is unclear whether CCL20 affects breast cancer progression by remodeling the tumor microenvironment (TME). Here, we observed that polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) were remarkably enriched in TME of CCL20-overexpressing cancer cell orthotopic allograft tumors. Mechanistically, CCL20 activated the differentiation of granulocyte-monocyte progenitors (GMPs) via its receptor C-C motif chemokine receptor 6 (CCR6) leading to the PMN-MDSC expansion. PMN-MDSCs from CCL20-overexpressing cell orthotopic allograft tumors (CCL20-modulated PMN-MDSCs) secreted amounts of C-X-C motif chemokine ligand 2 (CXCL2) and increased ALDH+ BCSCs via activating CXCR2/NOTCH1/HEY1 signaling pathway. Furthermore, C-X-C motif chemokine receptor 2 (CXCR2) antagonist SB225002 enhanced the docetaxel (DTX) effects on tumor growth by decreasing BCSCs in CCL20high-expressing tumors. These findings elucidated how CCL20 modulated the TME to promote cancer development, indicating a new therapeutic strategy by interfering with the interaction between PMN-MDSCs and BCSCs in breast cancer, especially in CCL20high-expressing breast cancer.


Sujet(s)
Tumeurs du sein , Chimiokines , Cellules myéloïdes suppressives , Cellules souches tumorales , Différenciation cellulaire , Ligands , Récepteurs à l'interleukine-8B , Humains , Animaux , Lignée cellulaire tumorale
11.
Cell ; 186(7): 1432-1447.e17, 2023 03 30.
Article de Anglais | MEDLINE | ID: mdl-37001503

RÉSUMÉ

Cancer immunotherapies, including adoptive T cell transfer, can be ineffective because tumors evolve to display antigen-loss-variant clones. Therapies that activate multiple branches of the immune system may eliminate escape variants. Here, we show that melanoma-specific CD4+ T cell therapy in combination with OX40 co-stimulation or CTLA-4 blockade can eradicate melanomas containing antigen escape variants. As expected, early on-target recognition of melanoma antigens by tumor-specific CD4+ T cells was required. Surprisingly, complete tumor eradication was dependent on neutrophils and partly dependent on inducible nitric oxide synthase. In support of these findings, extensive neutrophil activation was observed in mouse tumors and in biopsies of melanoma patients treated with immune checkpoint blockade. Transcriptomic and flow cytometry analyses revealed a distinct anti-tumorigenic neutrophil subset present in treated mice. Our findings uncover an interplay between T cells mediating the initial anti-tumor immune response and neutrophils mediating the destruction of tumor antigen loss variants.


Sujet(s)
Mélanome , Lymphocytes T , Souris , Animaux , Lymphocytes T/anatomopathologie , Granulocytes neutrophiles/anatomopathologie , Dérive et cassure antigéniques , Immunothérapie , Antigène CTLA-4
12.
Cancer Cell ; 41(3): 505-526, 2023 03 13.
Article de Anglais | MEDLINE | ID: mdl-36827980

RÉSUMÉ

Neutrophils are major effectors and regulators of the immune system. They play critical roles not only in the eradication of pathogens but also in cancer initiation and progression. Conversely, the presence of cancer affects neutrophil activity, maturation, and lifespan. By promoting or repressing key neutrophil functions, cancer cells co-opt neutrophil biology to their advantage. This co-opting includes hijacking one of neutrophils' most striking pathogen defense mechanisms: the formation of neutrophil extracellular traps (NETs). NETs are web-like filamentous extracellular structures of DNA, histones, and cytotoxic granule-derived proteins. Here, we discuss the bidirectional interplay by which cancer stimulates NET formation, and NETs in turn support disease progression. We review how vascular dysfunction and thrombosis caused by neutrophils and NETs underlie an elevated risk of death from cardiovascular events in cancer patients. Finally, we propose therapeutic strategies that may be effective in targeting NETs in the clinical setting.


Sujet(s)
Pièges extracellulaires , Tumeurs , Thrombose , Humains , Pièges extracellulaires/métabolisme , Granulocytes neutrophiles , Histone/métabolisme , Thrombose/étiologie , Thrombose/métabolisme , ADN/métabolisme , Tumeurs/métabolisme
13.
Nat Cell Biol ; 25(2): 298-308, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36658219

RÉSUMÉ

The EWS-FLI1 fusion oncoprotein deregulates transcription to initiate the paediatric cancer Ewing sarcoma. Here we used a domain-focused CRISPR screen to implicate the transcriptional repressor ETV6 as a unique dependency in this tumour. Using biochemical assays and epigenomics, we show that ETV6 competes with EWS-FLI1 for binding to select DNA elements enriched for short GGAA repeat sequences. Upon inactivating ETV6, EWS-FLI1 overtakes and hyper-activates these cis-elements to promote mesenchymal differentiation, with SOX11 being a key downstream target. We show that squelching of ETV6 with a dominant-interfering peptide phenocopies these effects and suppresses Ewing sarcoma growth in vivo. These findings reveal targeting of ETV6 as a strategy for neutralizing the EWS-FLI1 oncoprotein by reprogramming of genomic occupancy.


Sujet(s)
Sarcome d'Ewing , Enfant , Humains , Sarcome d'Ewing/génétique , Sarcome d'Ewing/métabolisme , Sarcome d'Ewing/anatomopathologie , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Protéine EWS de liaison à l'ARN/génétique , Protéine EWS de liaison à l'ARN/métabolisme , Protéine proto-oncogène c-fli-1/génétique , Protéine proto-oncogène c-fli-1/métabolisme , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme
14.
J Invest Surg ; 36(1): 2162170, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-36653180

RÉSUMÉ

OBJECTIVE: Cervical cancer is linked with the constitutive activation of growth factors and gene mutations-induced pro-survival signaling pathways. Herein, we purposed to explore the possible molecular mechanism of Foxo3a-mediated DNMT3B in the proliferation and migration of cervical cancer cells via mediating the PTEN promoter methylation. METHODS: Foxo3a expression in cervical cancer was tested by qRT-PCR and western blot experiments. The cervical cancer cell biological functions with overexpression of Foxo3a were evaluated by CCK-8 assay, Transwell experiment, and flow cytometry, respectively. MS-PCR was utilized for testing the PTEN methylation levels, and ChIP experiment was implemented for evaluating the enrichment of DNMT3B in the PTEN promoter region and the binding of Foxo3a and DNMT3B. The PTEN methylation and interference with Foxo3a expression were performed in cervical cancer cells, and then their impacts on cervical cancer cell biological functions were observed. RESULTS: FOXO3a was expressed at a low level in cervical cancer, and its overexpression contributed to a reduction in cell proliferative, migratory and invasive capabilities, and an elevation in apoptosis rate. Foxo3a blocked its methylation with the PTEN promoter by repressing DNMT3B activity. Upon treatment with methyltransferase inhibitor (5-aza-dc), the malignant phenotypes of cervical cancer cells were diminished. 5-aza-dc neutralized the impacts of silencing Foxo3a on malignant phenotypes. CONCLUSION: This research underlines that Foxo3a blocks its methylation with the PTEN promoter by inhibiting DNMT3B activity, which subsequently impedes cervical cancer cell progression.


Sujet(s)
Méthylation de l'ADN , Tumeurs du col de l'utérus , Humains , Femelle , Tumeurs du col de l'utérus/génétique , Transduction du signal , Azacitidine/métabolisme , Prolifération cellulaire/génétique , Régions promotrices (génétique) , Lignée cellulaire tumorale , Phosphohydrolase PTEN/génétique
15.
Food Chem ; 410: 135151, 2023 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-36623463

RÉSUMÉ

A triple-mode colorimetric and fluorescent sensing scheme based on manganese dioxide nanoparticles (MnO2NPs) and carbon quantum dots (CQDs) were developed to determine nitrite. MnO2NPs can oxidize 3,3',5,5'-tetramethylbenzidine (TMB) into a blue oxidation product (TMBox), which is further oxidized into a yellow diimine derivative by nitrite. The ratio of absorbance at 652 nm to 452 nm was monitored as signal response for UV-vis detection mode. A "turn-off" CQDs fluorescence probe was also constructed for fluorescent detection mode. Smartphone tool kit was used to capture the color of sample for smartphone-based measurement. Various analytical performance under different detection modes were obtained and compared. The proposed methods were applied to food samples with satisfactory recoveries (83.3-106 %). The results were validated with AOAC standard spectrophotometric method. The current triple-mode detection were accurate, convenient, low-cost and fast for analyzing nitrite in foods and water samples on-site.


Sujet(s)
Nanoparticules , Boîtes quantiques , Colorants fluorescents , Carbone , Nitrites , Colorimétrie/méthodes , Ordiphone , Composés du manganèse , Oxydes , Limite de détection
17.
Sci Rep ; 12(1): 14671, 2022 08 29.
Article de Anglais | MEDLINE | ID: mdl-36038588

RÉSUMÉ

This descriptive case series retrospectively reviewed medical records from thirty-one previously healthy, war-fighting veterans who self-reported exposure to airborne hazards while serving in Iraq and Afghanistan between 2003 and the present. They all noted new-onset dyspnea, which began during deployment or as a military contractor. Twenty-one subjects underwent non-invasive pulmonary diagnostic testing, including maximum expiratory pressure (MEP) and impulse oscillometry (IOS). In addition, five soldiers received a lung biopsy; tissue results were compared to a previously published sample from a soldier in our Iraq Afghanistan War Lung Injury database and others in our database with similar exposures, including burn pits. We also reviewed civilian control samples (5) from the Stony Brook University database. Military personnel were referred to our International Center of Excellence in Deployment Health and Medical Geosciences, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell under the auspices of Northwell IRB: 17-0140-FIMR Feinstein Institution for Medical Research "Clinicopathologic characteristics of Iraq Afghanistan War Lung Injury." We retrospectively examined medical records, including exposure data, radiologic imaging, and non-invasive pulmonary function testing (MGC Diagnostic Platinum Elite Plethysmograph) using the American Thoracic Society (ATS) standard interpretation based on Morgan et al., and for a limited cohort, biopsy data. Lung tissue, when available, was examined for carbonaceous particles, polycyclic aromatic hydrocarbons (Raman spectroscopy), metals, titanium connected to iron (Brookhaven National Laboratory, National Synchrotron Light Source II, Beamline 5-ID), oxidized metals, combustion temperature, inflammatory cell accumulation and fibrosis, neutrophil extracellular traps, Sirius red, Prussian Blue, as well as polarizable crystals/particulate matter/dust. Among twenty-one previously healthy, deployable soldiers with non-invasive pulmonary diagnostic tests, post-deployment, all had severely decreased MEP values, averaging 42% predicted. These same patients concurrently demonstrated abnormal airways reactance (X5Hz) and peripheral/distal airways resistance (D5-D20%) via IOS, averaging - 1369% and 23% predicted, respectively. These tests support the concept of airways hyperresponsiveness and distal airways narrowing, respectively. Among the five soldiers biopsied, all had constrictive bronchiolitis. We detected the presence of polycyclic aromatic hydrocarbons (PAH)-which are products of incomplete combustion-in the lung tissue of all five warfighters. All also had detectable titanium and iron in the lungs. Metals were all oxidized, supporting the concept of inhaling burned metals. Combustion temperature was consistent with that of burned petrol rather than higher temperatures noted with cigarettes. All were nonsmokers. Neutrophil extracellular traps were reported in two biopsies. Compared to our prior biopsies in our Middle East deployment database, these histopathologic results are similar, since all database biopsies have constrictive bronchiolitis, one has lung fibrosis with titanium bound to iron in fixed mathematical ratios of 1:7 and demonstrated polarizable crystals. These results, particularly constrictive bronchiolitis and polarizable crystals, support the prior data of King et al. (N. Engl. J. Med. 365:222-230, 2011) Soldiers in this cohort deployed to Iraq and Afghanistan since 2003, with exposure to airborne hazards, including sandstorms, burn pits, and improvised explosive devices, are at high risk for developing chronic clinical respiratory problems, including: (1) reduction in respiratory muscle strength; (2) airways hyperresponsiveness; and (3) distal airway narrowing, which may be associated with histopathologic evidence of lung damage, reflecting inhalation of burned particles from burn pits along with particulate matter/dust. Non-invasive pulmonary diagnostic tests are a predictor of burn pit-induced lung injury.


Sujet(s)
Bronchiolite oblitérante , Lésion pulmonaire , Hydrocarbures aromatiques polycycliques , Guerre d'Afghanistan 2001- , Afghanistan , Bronchiolite oblitérante/anatomopathologie , Poussière , Humains , Incinération , Iraq , Guerre d'Irak (2003-2011) , Fer , Poumon/anatomopathologie , Lésion pulmonaire/diagnostic , Lésion pulmonaire/étiologie , Lésion pulmonaire/anatomopathologie , Matière particulaire , Études rétrospectives , Titane , États-Unis/épidémiologie
18.
JCI Insight ; 7(14)2022 07 22.
Article de Anglais | MEDLINE | ID: mdl-35866483

RÉSUMÉ

Acute lung injury (ALI) can cause acute respiratory distress syndrome (ARDS), a lethal condition with limited treatment options and currently a common global cause of death due to COVID-19. ARDS secondary to transfusion-related ALI (TRALI) has been recapitulated preclinically by anti-MHC-I antibody administration to LPS-primed mice. In this model, we demonstrate that inhibitors of PTP1B, a protein tyrosine phosphatase that regulates signaling pathways of fundamental importance to homeostasis and inflammation, prevented lung injury and increased survival. Treatment with PTP1B inhibitors attenuated the aberrant neutrophil function that drives ALI and was associated with release of myeloperoxidase, suppression of neutrophil extracellular trap (NET) formation, and inhibition of neutrophil migration. Mechanistically, reduced signaling through the CXCR4 chemokine receptor, particularly to the activation of PI3Kγ/AKT/mTOR, was essential for these effects, linking PTP1B inhibition to promoting an aged-neutrophil phenotype. Considering that dysregulated activation of neutrophils has been implicated in sepsis and causes collateral tissue damage, we demonstrate that PTP1B inhibitors improved survival and ameliorated lung injury in an LPS-induced sepsis model and improved survival in the cecal ligation and puncture-induced (CLP-induced) sepsis model. Our data highlight the potential for PTP1B inhibition to prevent ALI and ARDS from multiple etiologies.


Sujet(s)
Lésion pulmonaire aigüe , COVID-19 , , Sepsie , Lésion pulmonaire aigüe/métabolisme , Animaux , Lipopolysaccharides/pharmacologie , Souris , Granulocytes neutrophiles , /étiologie , Sepsie/complications
19.
J Immunother Cancer ; 10(5)2022 05.
Article de Anglais | MEDLINE | ID: mdl-35613826

RÉSUMÉ

BACKGROUND: Although the antitumor efficacy of docetaxel (DTX) has long been attributed to the antimitotic activities, its impact on the tumor microenvironment (TME) has recently gained more attention. Macrophages are a major component of the TME and play a critical role in DTX efficacy; however, the underlying action mechanisms remain unclear. METHODS: DTX chemotherapeutic efficacy was demonstrated via both macrophage depletion and C-C motif chemokine ligand 3 (Ccl3)-knockout transgenic allograft mouse model. Ccl3-knockdown and Ccl3-overexpressing breast cancer cell allografts were used for the in vivo study. Combination therapy was used to evaluate the effect of Ccl3 induction on DTX chemosensitivity. Vital regulatory molecules and pathways were identified using RNA sequencing. Macrophage phagocytosis of cancer cells and its influence on cancer cell proliferation under DTX treatment were assessed using an in vitro coculture assay. Serum and tumor samples from patients with breast cancer were used to demonstrate the clinical relevance of our study. RESULTS: Our study revealed that Ccl3 induced by DTX in macrophages and cancer cells was indispensable for the chemotherapeutic efficacy of DTX. DTX-induced Ccl3 promoted proinflammatory macrophage polarization and subsequently facilitated phagocytosis of breast cancer cells and cancer stem cells. Ccl3 overexpression in cancer cells promoted proinflammatory macrophage polarization to suppress tumor progression and increase DTX chemosensitivity. Mechanistically, DTX induced Ccl3 by relieving the inhibition of cAMP-response element binding protein on Ccl3 via reactive oxygen species accumulation, and Ccl3 then promoted proinflammatory macrophage polarization via activation of the Ccl3-C-C motif chemokine receptor 5-p38/interferon regulatory factor 5 pathway. High CCL3 expression predicted better prognosis, and high CCL3 induction revealed better DTX chemosensitivity in patients with breast cancer. Furthermore, both the Creb inhibitor and recombinant mouse Ccl3 significantly enhanced DTX chemosensitivity. CONCLUSIONS: Our results indicate that Ccl3 induced by DTX triggers proinflammatory macrophage polarization and subsequently facilitates phagocytosis of cancer cells. Ccl3 induction in combination with DTX may provide a promising therapeutic rationale for increasing DTX chemosensitivity in breast cancer.


Sujet(s)
Tumeurs du sein , Chimiokine CCL3 , Macrophages , Animaux , Tumeurs du sein/immunologie , Tumeurs du sein/anatomopathologie , Prolifération cellulaire , Chimiokine CCL3/immunologie , Chimiokine CCL3/métabolisme , Docetaxel/pharmacologie , Docetaxel/usage thérapeutique , Femelle , Humains , Activation des macrophages , Macrophages/immunologie , Macrophages/anatomopathologie , Souris , Microenvironnement tumoral
20.
PLoS One ; 17(5): e0267798, 2022.
Article de Anglais | MEDLINE | ID: mdl-35511773

RÉSUMÉ

Several studies have demonstrated the association of lncRNAs with a variety of cancers. Here, we explored the role of LINC00922 in gastric cancer (GC) using bioinformatics approaches and in vitro experiments. We examined the expression of LINC00922 and the prognosis of GC patients based on data from The Cancer Genome Atlas (TCGA) and Gene Expression Profiling Interactive Analysis (GEPIA). LINC00922-related genes were identified by the Multi Experiment Matrix (MEM) database and The Atlas of Noncoding RNAs in Cancer (TANRIC), followed by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and protein-protein interaction analysis. The significance of LINC00922 in cell proliferation, apoptosis, invasion and migration was assessed by MTT assay, flow cytometry, Transwell assay and wound-healing assay. The expression of LINC00922 was increased in GC tissues compared with adjacent non-tumor tissues, and increased LINC00922 expression was correlated with poor overall survival and disease-free survival. In addition, 336 overlapping genes were identified by the MEM database and TANRIC and found to be involved in GC-related biological processes, such as cell adhesion and migration, as well as TGF-ß signaling. In the protein-protein interaction network, hub genes, such as FSTL3 and LAMC1, were identified. LINC00922 overexpression significantly promoted cell proliferation and invasion in vitro, whereas LINC00922 knockdown exerted opposite effects. In summary, our findings indicate that LINC00922 is overexpressed in GC tissues, suggesting that it might play a role in the development and progression of GC, and thus, it might serve as a prognostic indicator of GC.


Sujet(s)
Protéines apparentées à la follistatine , ARN long non codant , Tumeurs de l'estomac , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Protéines apparentées à la follistatine/génétique , Régulation de l'expression des gènes tumoraux , Humains , Pronostic , ARN long non codant/génétique , ARN long non codant/métabolisme , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE