Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
PLoS One ; 19(6): e0304522, 2024.
Article in English | MEDLINE | ID: mdl-38837993

ABSTRACT

BACKGROUND: A subset of individuals (10-20%) experience post-COVID condition (PCC) subsequent to initial SARS-CoV-2 infection, which lacks effective treatment. PCC carries a substantial global burden associated with negative economic and health impacts. This study aims to evaluate the association between plasma taurine levels with self-reported symptoms and adverse clinical outcomes in patients with PCC. METHODS AND FINDINGS: We analyzed the plasma proteome and metabolome of 117 individuals during their acute COVID-19 hospitalization and at the convalescence phase six-month post infection. Findings were compared with 28 age and sex-matched healthy controls. Plasma taurine levels were negatively associated with PCC symptoms and correlated with markers of inflammation, tryptophan metabolism, and gut dysbiosis. Stratifying patients based on the trajectories of plasma taurine levels during six-month follow-up revealed a significant association with adverse clinical events. Increase in taurine levels during the transition to convalescence were associated with a reduction in adverse events independent of comorbidities and acute COVID-19 severity. In a multivariate analysis, increased plasma taurine level between acute and convalescence phase was associated with marked protection from adverse clinical events with a hazard ratio of 0.13 (95% CI: 0.05-0.35; p<0.001). CONCLUSIONS: Taurine emerges as a promising predictive biomarker and potential therapeutic target in PCC. Taurine supplementation has already demonstrated clinical benefits in various diseases and warrants exploration in large-scale clinical trials for alleviating PCC.


Subject(s)
COVID-19 , SARS-CoV-2 , Taurine , Humans , Taurine/blood , COVID-19/blood , COVID-19/complications , Female , Male , Middle Aged , SARS-CoV-2/isolation & purification , Adult , Biomarkers/blood , Aged , Post-Acute COVID-19 Syndrome , Case-Control Studies , Metabolome , Symptom Burden
2.
Nat Commun ; 15(1): 4259, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38769334

ABSTRACT

Tools for predicting COVID-19 outcomes enable personalized healthcare, potentially easing the disease burden. This collaborative study by 15 institutions across Europe aimed to develop a machine learning model for predicting the risk of in-hospital mortality post-SARS-CoV-2 infection. Blood samples and clinical data from 1286 COVID-19 patients collected from 2020 to 2023 across four cohorts in Europe and Canada were analyzed, with 2906 long non-coding RNAs profiled using targeted sequencing. From a discovery cohort combining three European cohorts and 804 patients, age and the long non-coding RNA LEF1-AS1 were identified as predictive features, yielding an AUC of 0.83 (95% CI 0.82-0.84) and a balanced accuracy of 0.78 (95% CI 0.77-0.79) with a feedforward neural network classifier. Validation in an independent Canadian cohort of 482 patients showed consistent performance. Cox regression analysis indicated that higher levels of LEF1-AS1 correlated with reduced mortality risk (age-adjusted hazard ratio 0.54, 95% CI 0.40-0.74). Quantitative PCR validated LEF1-AS1's adaptability to be measured in hospital settings. Here, we demonstrate a promising predictive model for enhancing COVID-19 patient management.


Subject(s)
COVID-19 , Hospital Mortality , Machine Learning , RNA, Long Noncoding , SARS-CoV-2 , Humans , COVID-19/mortality , COVID-19/virology , COVID-19/genetics , Male , Female , Aged , RNA, Long Noncoding/genetics , Middle Aged , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Europe/epidemiology , Canada/epidemiology , Cohort Studies , Aged, 80 and over , Adult
3.
Am J Respir Crit Care Med ; 209(12): 1453-1462, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38324627

ABSTRACT

Rationale: Pseudomonas aeruginosa is the major bacterial pathogen colonizing the airways of adult patients with cystic fibrosis (CF) and causes chronic infections that persist despite antibiotic therapy. Intracellular bacteria may represent an unrecognized reservoir of bacteria that evade the immune system and antibiotic therapy. Although the ability of P. aeruginosa to invade and survive within epithelial cells has been described in vitro in different epithelial cell models, evidence of this intracellular lifestyle in human lung tissues is currently lacking. Objectives: To detect and characterize intracellular P. aeruginosa in CF airway epithelium from human lung explant tissues. Methods: We sampled lung explant tissues from patients with CF undergoing lung transplantation and non-CF lung donor control tissue. We analyzed lung tissue sections for the presence of intracellular P. aeruginosa using quantitative culture and microscopy, in parallel to histopathology and airway morphometry. Measurements and Main Results: P. aeruginosa was isolated from the lungs of seven patients with CF undergoing lung transplantation. Microscopic assessment revealed the presence of intracellular P. aeruginosa within airway epithelial cells in three of the seven patients analyzed at a varying but low frequency. We observed those events occurring in lung regions with high bacterial burden. Conclusions: This is the first study describing the presence of intracellular P. aeruginosa in CF lung tissues. Although intracellular P. aeruginosa in airway epithelial cells is likely relatively rare, our findings highlight the plausible occurrence of this intracellular bacterial reservoir in chronic CF infections.


Subject(s)
Cystic Fibrosis , Lung Transplantation , Lung , Pseudomonas Infections , Pseudomonas aeruginosa , Respiratory Mucosa , Humans , Cystic Fibrosis/microbiology , Cystic Fibrosis/complications , Female , Male , Adult , Respiratory Mucosa/microbiology , Respiratory Mucosa/pathology , Pseudomonas Infections/microbiology , Lung/microbiology , Lung/pathology , Young Adult , Epithelial Cells/microbiology
4.
Vascul Pharmacol ; 154: 107250, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38043758

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus has been at the forefront of health sciences research since its emergence in China in 2019 that quickly led to a global pandemic. As a result of this research, and the large numbers of infected patients globally, there were rapid enhancements made in our understanding of Coronavirus disease 2019 (COVID-19) pathology, including its role in the development of uncontrolled immune responses and its link to the development of endotheliitis and endothelial dysfunction. There were also some noted differences in the rate and severity of infection between males and females with acute COVID. Some individuals infected with SARS-CoV-2 also experience long-COVID, an important hallmark symptom of this being Myalgic Encephalomyelitis-Chronic Fatigue Syndrome (ME-CFS), also experienced differently between males and females. The purpose of this review is to discuss the impact of sex on the vasculature during acute and long COVID-19, present any link between ME-CFS and endothelial dysfunction, and provide evidence for the relationship between ME-CFS and the immune system. We also will delineate biological sex differences observed in other post viral infections and, assess if sex differences exist in how the immune system responds to viral infection causing ME-CFS.


Subject(s)
COVID-19 , Fatigue Syndrome, Chronic , Vascular Diseases , Humans , Female , Male , Post-Acute COVID-19 Syndrome , SARS-CoV-2 , Fatigue Syndrome, Chronic/epidemiology , Sex Characteristics
5.
Clin Proteomics ; 20(1): 44, 2023 Oct 24.
Article in English | MEDLINE | ID: mdl-37875801

ABSTRACT

The quest for understanding and managing the long-term effects of COVID-19, often referred to as Long COVID or post-COVID-19 condition (PCC), remains an active research area. Recent findings highlighted angiopoietin-1 (ANG-1) and p-selectin (P-SEL) as potential diagnostic markers, but validation is essential, given the inconsistency in COVID-19 biomarker studies. Leveraging the biobanque québécoise de la COVID-19 (BQC19) biobank, we analyzed the data of 249 participants. Both ANG-1 and P-SEL levels were significantly higher in patients with PCC participants compared with control subjects at 3 months using the Mann-Whitney U test. We managed to reproduce and validate the findings, emphasizing the importance of collaborative biobanking efforts in enhancing the reproducibility and credibility of Long COVID research outcomes.

6.
Am J Rhinol Allergy ; 37(6): 638-645, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37475202

ABSTRACT

BACKGROUND: Chronic rhinosinusitis with nasal polyposis (CRSwNP) is a multifactorial disease with no known single cause, but it is thought that bacteria play a role in the disease process. OBJECTIVE: This pilot study aims to assess the longitudinal effect of corticosteroid therapy on sinus microbiota in chronic rhinosinusitis patients with nasal polyposis (CRSwNP). METHODS: A longitudinal prospective case-control study was done on patients with CRSwNP and healthy controls. Patients with CRSwNP were randomly allocated to a corticosteroids and antibiotics treatment group (CRSwNP-SA) or a corticosteroid-only treatment group (CRSwNP-S). Data were collected at three-time points (before treatment, 1, and 3 months after treatment). Specimens were cultured and matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry (MS) was used as a bacterial detection method. RESULTS: Data from 29 patients with CRSwNP (16 CRSwNP-SA and 13 CRSwNP-S) was compared to 15 healthy subjects. Patients reported significant symptom improvement initially (1 month), but not in the long-term (3 months). This result was found in both treatment groups, whether or not antibiotics were used. After 3 months from treatment, the prevalence of Corynebacterium genera tended to increase in the CRSwNP-SA, while Staphylococcus and Gram-negative genera (Pseudomonas) tended to increase in the CRSwNP-S. Smoking, aspirin sensitivity, and previous endoscopic sinus surgery were found to be co-factors significantly associated with the response to systemic corticosteroid therapy. CONCLUSION: In this pilot study, both treatment options were effective to improve symptoms in the short-term but not in the long-term, and were not linked to any clear sinus microbiota response. As a result, this study supports the avoidance of systemic antibiotics without evidence of active infection.


Subject(s)
Microbiota , Nasal Polyps , Rhinitis , Sinusitis , Humans , Case-Control Studies , Pilot Projects , Rhinitis/complications , Sinusitis/complications , Adrenal Cortex Hormones/therapeutic use , Nasal Polyps/drug therapy , Nasal Polyps/complications , Chronic Disease , Anti-Bacterial Agents/therapeutic use
8.
Clin Exp Metastasis ; 39(2): 323-333, 2022 04.
Article in English | MEDLINE | ID: mdl-34767138

ABSTRACT

Surgical resection, the cornerstone of curative intent treatment for gastric adenocarcinoma, is associated with a high rate of infection-related post-operative complications, leading to an increased incidence of metastasis to the peritoneum. However, the mechanisms underlying this process are poorly understood. Lipopolysaccharide (LPS), an antigen from Gram-negative bacteria, represents a potential mechanism via induction of local and systemic inflammation through activation of Toll-like receptor (TLR). Here, we use both a novel ex vivo model of peritoneal metastasis and in vivo animal models to assess gastric cancer cell adhesion to peritoneum both before and after inhibition of the TLR4 pathway. We demonstrate that activation of TLR4 by either LPS or Gram-negative bacteria (E. coli) significantly increases the adherence of gastric cancer cells to human peritoneal mesothelial cells, and that this increased adherence is abrogated by inhibition of the TLR4 signal cascade and downstream TAK1 and MEK1/2 pathways. We also demonstrate that the influence of LPS on adherence extends to peritoneal tissue and metastatic spread. Furthermore, we show that loss of TLR4 at the site of metastasis reduces tumor cell adhesion, implicating the TLR4 signaling cascade in potentiating metastatic adhesion and peritoneal spread. These results identify potential therapeutic targets for the clinical management of patients undergoing resection for gastric cancer.


Subject(s)
Adenocarcinoma , Peritoneal Neoplasms , Stomach Neoplasms , Animals , Escherichia coli/metabolism , Humans , Lipopolysaccharides/pharmacology , Peritoneum , Toll-Like Receptor 4/metabolism
9.
Sci Rep ; 11(1): 23928, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34907210

ABSTRACT

Identification of transcriptional regulatory mechanisms and signaling networks involved in the response of host cells to infection by SARS-CoV-2 is a powerful approach that provides a systems biology view of gene expression programs involved in COVID-19 and may enable the identification of novel therapeutic targets and strategies to mitigate the impact of this disease. In this study, our goal was to identify a transcriptional regulatory network that is associated with gene expression changes between samples infected by SARS-CoV-2 and those that are infected by other respiratory viruses to narrow the results on those enriched or specific to SARS-CoV-2. We combined a series of recently developed computational tools to identify transcriptional regulatory mechanisms involved in the response of epithelial cells to infection by SARS-CoV-2, and particularly regulatory mechanisms that are specific to this virus when compared to other viruses. In addition, using network-guided analyses, we identified kinases associated with this network. The results identified pathways associated with regulation of inflammation (MAPK14) and immunity (BTK, MBX) that may contribute to exacerbate organ damage linked with complications of COVID-19. The regulatory network identified herein reflects a combination of known hits and novel candidate pathways supporting the novel computational pipeline presented herein to quickly narrow down promising avenues of investigation when facing an emerging and novel disease such as COVID-19.


Subject(s)
COVID-19/genetics , Gene Expression Profiling/methods , SARS-CoV-2/pathogenicity , Sequence Analysis, RNA/methods , A549 Cells , Cell Line , Epithelial Cells/chemistry , Epithelial Cells/cytology , Epithelial Cells/virology , Gene Expression Regulation , Humans , Models, Biological , Systems Biology
10.
Biochemistry ; 60(50): 3829-3840, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34845903

ABSTRACT

Catalytic promiscuity is the coincidental ability to catalyze nonbiological reactions in the same active site as the native biological reaction. Several lines of evidence show that catalytic promiscuity plays a role in the evolution of new enzyme functions. Thus, studying catalytic promiscuity can help identify structural features that predispose an enzyme to evolve new functions. This study identifies a potentially preadaptive residue in a promiscuous N-succinylamino acid racemase/o-succinylbenzoate synthase (NSAR/OSBS) enzyme from Amycolatopsis sp. T-1-60. This enzyme belongs to a branch of the OSBS family which includes many catalytically promiscuous NSAR/OSBS enzymes. R266 is conserved in all members of the NSAR/OSBS subfamily. However, the homologous position is usually hydrophobic in other OSBS subfamilies, whose enzymes lack NSAR activity. The second-shell amino acid R266 is close to the catalytic acid/base K263, but it does not contact the substrate, suggesting that R266 could affect the catalytic mechanism. Mutating R266 to glutamine in Amycolatopsis NSAR/OSBS profoundly reduces NSAR activity but moderately reduces OSBS activity. This is due to a 1000-fold decrease in the rate of proton exchange between the substrate and the general acid/base catalyst K263. This mutation is less deleterious for the OSBS reaction because K263 forms a cation-π interaction with the OSBS substrate and/or the intermediate, rather than acting as a general acid/base catalyst. Together, the data explain how R266 contributes to NSAR reaction specificity and was likely an essential preadaptation for the evolution of NSAR activity.


Subject(s)
Amino Acid Isomerases/chemistry , Amino Acid Isomerases/metabolism , Carbon-Carbon Lyases/chemistry , Carbon-Carbon Lyases/metabolism , Amino Acid Isomerases/genetics , Amino Acid Sequence , Amino Acid Substitution , Amycolatopsis/enzymology , Amycolatopsis/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biocatalysis , Carbon-Carbon Lyases/genetics , Catalytic Domain/genetics , Conserved Sequence , Crystallography, X-Ray , Enzyme Stability/genetics , Evolution, Molecular , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
11.
Anal Bioanal Chem ; 413(20): 5135-5146, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34173039

ABSTRACT

Drug safety assessment in the early phases of drug discovery is critical to facilitate the rapid development of novel therapeutics. Recently, teleost zebrafish (Danio rerio) has emerged as a promising vertebrate model for the assessment of drug safety. Zebrafish is a convenient model because of its small size, high fecundity, embryo transparency, and ex utero development. In this study, we developed a matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) method applied to zebrafish larvae to investigate safety and metabolism of sahaquine (Sq), an anticancer agent inhibiting histone deacetylase 6. This technique improves on prior studies using liquid chromatography-mass spectrometry (LC-MS) by adding analysis of the drug spatial distribution. Using this method, it was determined that Sq dissolved in fish water (1-2000 µM) did not reach the larval body and was mainly distributed throughout the yolk. High Sq concentration (800 µM) administered intravenously allowed the compound to reach the larval body but did not induce phenotypic abnormalities. Sq was metabolized into its glucuronidated form within 24 h and was excreted within 72 h. MALDI MSI was instrumental in showing that Sq-glucuronide was mainly formed in the gut and slightly in yolk syncytial layer, and provided valuable insights into xenobiotics elimination in zebrafish larvae. This study indicates that Sq has a good safety profile and merits further investigations in other disease models. In addition, the optimized MALDI MSI protocol provided here can be widely applied to study distribution and metabolic fate of other structurally related molecules.


Subject(s)
Mass Spectrometry/methods , Animals , Cell Line, Tumor , Embryo, Nonmammalian/drug effects , Humans , Larva/drug effects , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Zebrafish
12.
PLoS One ; 16(5): e0245031, 2021.
Article in English | MEDLINE | ID: mdl-34010280

ABSTRACT

SARS-CoV-2 infection causing the novel coronavirus disease 2019 (COVID-19) has been responsible for more than 2.8 million deaths and nearly 125 million infections worldwide as of March 2021. In March 2020, the World Health Organization determined that the COVID-19 outbreak is a global pandemic. The urgency and magnitude of this pandemic demanded immediate action and coordination between local, regional, national, and international actors. In that mission, researchers require access to high-quality biological materials and data from SARS-CoV-2 infected and uninfected patients, covering the spectrum of disease manifestations. The "Biobanque québécoise de la COVID-19" (BQC19) is a pan-provincial initiative undertaken in Québec, Canada to enable the collection, storage and sharing of samples and data related to the COVID-19 crisis. As a disease-oriented biobank based on high-quality biosamples and clinical data of hospitalized and non-hospitalized SARS-CoV-2 PCR positive and negative individuals. The BQC19 follows a legal and ethical management framework approved by local health authorities. The biosamples include plasma, serum, peripheral blood mononuclear cells and DNA and RNA isolated from whole blood. In addition to the clinical variables, BQC19 will provide in-depth analytical data derived from the biosamples including whole genome and transcriptome sequencing, proteome and metabolome analyses, multiplex measurements of key circulating markers as well as anti-SARS-CoV-2 antibody responses. BQC19 will provide the scientific and medical communities access to data and samples to better understand, manage and ultimately limit, the impact of COVID-19. In this paper we present BQC19, describe the process according to which it is governed and organized, and address opportunities for future research collaborations. BQC19 aims to be a part of a global communal effort addressing the challenges of COVID-19.


Subject(s)
Biological Specimen Banks/organization & administration , COVID-19/pathology , COVID-19/epidemiology , COVID-19/genetics , COVID-19/metabolism , Humans , Information Dissemination/methods , Pandemics , Quebec/epidemiology , SARS-CoV-2/isolation & purification
13.
PLoS Pathog ; 17(3): e1009375, 2021 03.
Article in English | MEDLINE | ID: mdl-33690714

ABSTRACT

Pseudomonas aeruginosa causes chronic airway infections, a major determinant of lung inflammation and damage in cystic fibrosis (CF). Loss-of-function lasR mutants commonly arise during chronic CF infections, are associated with accelerated lung function decline in CF patients and induce exaggerated neutrophilic inflammation in model systems. In this study, we investigated how lasR mutants modulate airway epithelial membrane bound ICAM-1 (mICAM-1), a surface adhesion molecule, and determined its impact on neutrophilic inflammation in vitro and in vivo. We demonstrated that LasR-deficient strains induce increased mICAM-1 levels in airway epithelial cells compared to wild-type strains, an effect attributable to the loss of mICAM-1 degradation by LasR-regulated proteases and associated with enhanced neutrophil adhesion. In a subacute airway infection model, we also observed that lasR mutant-infected mice displayed greater airway epithelial ICAM-1 expression and increased neutrophilic pulmonary inflammation. Our findings provide new insights into the intricate interplay between lasR mutants, LasR-regulated proteases and airway epithelial ICAM-1 expression, and reveal a new mechanism involved in the exaggerated inflammatory response induced by lasR mutants.


Subject(s)
Cystic Fibrosis/complications , Pneumonia/microbiology , Pseudomonas aeruginosa/pathogenicity , Respiratory System/parasitology , Animals , Bacterial Proteins/metabolism , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Gene Expression Regulation, Bacterial/physiology , Humans , Mice , Pneumonia/complications , Pseudomonas Infections/immunology , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/metabolism , Respiratory System/metabolism , Trans-Activators/genetics
14.
PLoS Comput Biol ; 17(3): e1008810, 2021 03.
Article in English | MEDLINE | ID: mdl-33684134

ABSTRACT

Abnormal coagulation and an increased risk of thrombosis are features of severe COVID-19, with parallels proposed with hemophagocytic lymphohistiocytosis (HLH), a life-threating condition associated with hyperinflammation. The presence of HLH was described in severely ill patients during the H1N1 influenza epidemic, presenting with pulmonary vascular thrombosis. We tested the hypothesis that genes causing primary HLH regulate pathways linking pulmonary thromboembolism to the presence of SARS-CoV-2 using novel network-informed computational algorithms. This approach led to the identification of Neutrophils Extracellular Traps (NETs) as plausible mediators of vascular thrombosis in severe COVID-19 in children and adults. Taken together, the network-informed analysis led us to propose the following model: the release of NETs in response to inflammatory signals acting in concert with SARS-CoV-2 damage the endothelium and direct platelet-activation promoting abnormal coagulation leading to serious complications of COVID-19. The underlying hypothesis is that genetic and/or environmental conditions that favor the release of NETs may predispose individuals to thrombotic complications of COVID-19 due to an increase risk of abnormal coagulation. This would be a common pathogenic mechanism in conditions including autoimmune/infectious diseases, hematologic and metabolic disorders.


Subject(s)
COVID-19/complications , COVID-19/genetics , Extracellular Traps/genetics , Lymphohistiocytosis, Hemophagocytic/complications , Lymphohistiocytosis, Hemophagocytic/genetics , Models, Biological , SARS-CoV-2/genetics , Thrombosis/etiology , Thrombosis/genetics , Algorithms , Cell Degranulation/genetics , Computational Biology , Gene Expression Regulation , Gene Regulatory Networks , Genetic Predisposition to Disease , Humans , Pandemics , Protein Interaction Maps , Pulmonary Embolism/etiology , Pulmonary Embolism/genetics , Viral Proteins/genetics
15.
Nat Commun ; 11(1): 6092, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33257709

ABSTRACT

The approval of bedaquiline (BDQ) for the treatment of tuberculosis has generated substantial interest in inhibiting energy metabolism as a therapeutic paradigm. However, it is not known precisely how BDQ triggers cell death in Mycobacterium tuberculosis (Mtb). Using 13C isotopomer analysis, we show that BDQ-treated Mtb redirects central carbon metabolism to induce a metabolically vulnerable state susceptible to genetic disruption of glycolysis and gluconeogenesis. Metabolic flux profiles indicate that BDQ-treated Mtb is dependent on glycolysis for ATP production, operates a bifurcated TCA cycle by increasing flux through the glyoxylate shunt, and requires enzymes of the anaplerotic node and methylcitrate cycle. Targeting oxidative phosphorylation (OXPHOS) with BDQ and simultaneously inhibiting substrate level phosphorylation via genetic disruption of glycolysis leads to rapid sterilization. Our findings provide insight into the metabolic mechanism of BDQ-induced cell death and establish a paradigm for the development of combination therapies that target OXPHOS and glycolysis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Diarylquinolines/pharmacology , Glycolysis/drug effects , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Antitubercular Agents/pharmacology , Bacterial Proteins/metabolism , Carbon Cycle/drug effects , Citric Acid Cycle/drug effects , Energy Metabolism/drug effects , Glyoxylates , Mycobacterium tuberculosis/genetics , Oxidative Phosphorylation , Tuberculosis/microbiology
17.
J Law Biosci ; 7(1): lsaa020, 2020.
Article in English | MEDLINE | ID: mdl-32728465

ABSTRACT

Effective responses to the COVID-19 pandemic require novel solutions for research and responsible data sharing. Biobanking presents itself as a key priority in furthering our understanding of COVID-19. In this article, we propose a tripartite approach to consent to create resources for research relating to COVID-19. The approach aims to link three levels of participation: COVID-19 patients, respiratory/infectious disease patients, and longitudinal study participants. We explore the potential approaches that can be taken to consent processes with these three participant groups. We furthermore describe an access model for both single-site and multi-site data and sample storage. Through dealing with these topics at a high level, the model may be adapted to local legal and ethical requirements while still pursuing its ultimate goal: the creation of a research infrastructure that supports transparent, strong, and open science.

18.
J Exp Med ; 217(6)2020 06 01.
Article in English | MEDLINE | ID: mdl-32302401

ABSTRACT

Coronavirus disease 2019 (COVID-19) is a novel, viral-induced respiratory disease that in ∼10-15% of patients progresses to acute respiratory distress syndrome (ARDS) triggered by a cytokine storm. In this Perspective, autopsy results and literature are presented supporting the hypothesis that a little known yet powerful function of neutrophils-the ability to form neutrophil extracellular traps (NETs)-may contribute to organ damage and mortality in COVID-19. We show lung infiltration of neutrophils in an autopsy specimen from a patient who succumbed to COVID-19. We discuss prior reports linking aberrant NET formation to pulmonary diseases, thrombosis, mucous secretions in the airways, and cytokine production. If our hypothesis is correct, targeting NETs directly and/or indirectly with existing drugs may reduce the clinical severity of COVID-19.


Subject(s)
Betacoronavirus , Coronavirus Infections/pathology , Extracellular Traps , Lung Diseases , Neutrophils/pathology , Pneumonia, Viral/pathology , COVID-19 , Coronavirus Infections/complications , Cytokines/metabolism , Humans , Lung Diseases/etiology , Lung Diseases/metabolism , Lung Diseases/pathology , Pandemics , Pneumonia, Viral/complications , SARS-CoV-2
19.
J Immunol ; 204(8): 2285-2294, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32169849

ABSTRACT

Neutrophils promote tumor growth and metastasis at multiple stages of cancer progression. One mechanism through which this occurs is via release of neutrophil extracellular traps (NETs). We have previously shown that NETs trap tumor cells in both the liver and the lung, increasing their adhesion and metastasis following postoperative complications. Multiple studies have since shown that NETs play a role in tumor progression and metastasis. NETs are composed of nuclear DNA-derived web-like structures decorated with neutrophil-derived proteins. However, it is unknown which, if any, of these NET-affiliated proteins is responsible for inducing the metastatic phenotype. In this study, we identify the NET-associated carcinoembryonic Ag cell adhesion molecule 1 (CEACAM1) as an essential element for this interaction. Indeed, blocking CEACAM1 on NETs, or knocking it out in a murine model, leads to a significant decrease in colon carcinoma cell adhesion, migration and metastasis. Thus, this work identifies NET-associated CEACAM1 as a putative therapeutic target to prevent the metastatic progression of colon carcinoma.


Subject(s)
Antigens, CD/metabolism , Carcinoembryonic Antigen/metabolism , Cell Adhesion Molecules/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Extracellular Traps/immunology , Extracellular Traps/metabolism , Neutrophils/immunology , A549 Cells , Animals , Cell Line, Tumor , Colonic Neoplasms/immunology , HT29 Cells , Humans , Mice , Neutrophils/pathology
20.
Protein Cell ; 11(3): 187-201, 2020 03.
Article in English | MEDLINE | ID: mdl-31956962

ABSTRACT

While emerging data suggest nucleotide oligomerization domain receptor 1 (NOD1), a cytoplasmic pattern recognition receptor, may play an important and complementary role in the immune response to bacterial infection, its role in cancer metastasis is entirely unknown. Hence, we sought to determine the effects of NOD1 on metastasis. NOD1 expression in paired human primary colon cancer, human and murine colon cancer cells were determined using immunohistochemistry and immunoblotting (WB). Clinical significance of NOD1 was assessed using TCGA survival data. A series of in vitro and in vivo functional assays, including adhesion, migration, and metastasis, was conducted to assess the effect of NOD1. C12-iE-DAP, a highly selective NOD1 ligand derived from gram-negative bacteria, was used to activate NOD1. ML130, a specific NOD1 inhibitor, was used to block C12-iE-DAP stimulation. Stable knockdown (KD) of NOD1 in human colon cancer cells (HT29) was constructed with shRNA lentiviral transduction and the functional assays were thus repeated. Lastly, the predominant signaling pathway of NOD1-activation was identified using WB and functional assays in the presence of specific kinase inhibitors. Our data demonstrate that NOD1 is highly expressed in human colorectal cancer (CRC) and human and murine CRC cell lines. Clinically, we demonstrate that this increased NOD1 expression negatively impacts survival in patients with CRC. Subsequently, we identify NOD1 activation by C12-iE-DAP augments CRC cell adhesion, migration and metastasis. These effects are predominantly mediated via the p38 mitogen activated protein kinase (MAPK) pathway. This is the first study implicating NOD1 in cancer metastasis, and thus identifying this receptor as a putative therapeutic target.


Subject(s)
Adenocarcinoma/metabolism , Colonic Neoplasms/metabolism , Nod1 Signaling Adaptor Protein/physiology , Adenocarcinoma/pathology , Animals , Cell Adhesion , Cell Line , Cell Movement , Colonic Neoplasms/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...