Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Emerg Infect Dis ; 29(5): 1007-1010, 2023 05.
Article in English | MEDLINE | ID: covidwho-20245370

ABSTRACT

Increasing reports of invasive Streptococcus pyogenes infections mandate surveillance for toxigenic lineage M1UK. An allele-specific PCR was developed to distinguish M1UK from other emm1 strains. The M1UK lineage represented 91% of invasive emm1 isolates in England in 2020. Allele-specific PCR will permit surveillance for M1UK without need for genome sequencing.


Subject(s)
Scarlet Fever , Streptococcal Infections , Humans , Streptococcus pyogenes/genetics , Scarlet Fever/epidemiology , Alleles , England/epidemiology , Streptococcal Infections/diagnosis , Streptococcal Infections/epidemiology , Polymerase Chain Reaction , Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/genetics
2.
Microb Genom ; 9(6)2023 06.
Article in English | MEDLINE | ID: covidwho-20244144

ABSTRACT

Invasive group A streptococcal (iGAS) disease cases increased in the first half of 2022 in the Netherlands, with a remarkably high proportion of emm4 isolates. Whole-genome sequence analysis of 66 emm4 isolates, 40 isolates from the pre-coronavirus disease 2019 (COVID-19) pandemic period 2009-2019 and 26 contemporary isolates from 2022, identified a novel Streptococcus pyogenes lineage (M4NL22), which accounted for 85 % of emm4 iGAS cases in 2022. Surprisingly, we detected few isolates of the emm4 hypervirulent clone, which has replaced nearly all other emm4 in the USA and the UK. M4NL22 displayed genetic differences compared to other emm4 strains, although these were of unclear biological significance. In publicly available data, we identified a single Norwegian isolate belonging to M4NL22, which was sampled after the isolates from this study, possibly suggesting export of M4NL22 to Norway. In conclusion, our study identified a novel S. pyogenes emm4 lineage underlying an increase of iGAS disease in early 2022 in the Netherlands and the results have been promptly communicated to public health officials.


Subject(s)
COVID-19 , Streptococcal Infections , Humans , Antigens, Bacterial/genetics , Netherlands/epidemiology , Bacterial Outer Membrane Proteins/genetics , Carrier Proteins/genetics , Streptococcal Infections/epidemiology , Streptococcus pyogenes/genetics
3.
Int J Infect Dis ; 130 Suppl 1: S47-S51, 2023 May.
Article in English | MEDLINE | ID: covidwho-2298476

ABSTRACT

OBJECTIVES: Despite concerted efforts, Mycobacterium tuberculosis (M.tb), the pathogen that causes tuberculosis (TB), continues to be a burden on global health, regaining its dubious distinction in 2022 as the world's biggest infectious killer with global COVID-19 deaths steadily declining. The complex nature of M.tb, coupled with different pathogenic stages, has highlighted the need for the development of novel immunization approaches to combat this ancient infectious agent. Intensive efforts over the last couple of decades have identified alternative approaches to improve upon traditional vaccines that are based on killed pathogens, live attenuated agents, or subunit recombinant antigens formulated with adjuvants. Massive funding and rapid advances in RNA-based vaccines for immunization have recently transformed the possibility of protecting global populations from viral pathogens, such as SARS-CoV-2. Similar efforts to combat bacterial pathogens such as M.tb have been significantly slower to implement. METHODS: In this review, we discuss the application of a novel replicating RNA (repRNA)-based vaccine formulated and delivered in nanostructured lipids. RESULTS: Our preclinical data are the first to report that RNA platforms are a viable system for TB vaccines and should be pursued with high-priority M.tb antigens containing cluster of differentiation (CD4+) and CD8+ T-cell epitopes. CONCLUSION: This RNA vaccine shows promise for use against intracellular bacteria such as M.tb as demonstrated by the feasibility of construction, enhanced induction of cell-mediated and humoral immune responses, and improved bacterial burden outcomes in in vivo aerosol-challenged preclinical TB models.


Subject(s)
COVID-19 , Mycobacterium tuberculosis , Tuberculosis Vaccines , Tuberculosis , Humans , COVID-19/prevention & control , SARS-CoV-2 , Tuberculosis/prevention & control , Mycobacterium tuberculosis/genetics , Antigens, Bacterial
4.
Int J Mol Sci ; 24(7)2023 Mar 24.
Article in English | MEDLINE | ID: covidwho-2305215

ABSTRACT

The presentation of viral antigens on nanoparticles in multivalent arrays has emerged as a valuable technology for vaccines. On the nanoparticle surface, highly ordered, repetitive arrays of antigens can mimic their geometric arrangement on virion surfaces and elicit stronger humoral responses than soluble viral antigens. More recently, bacterial antigens have been presented on self-assembling protein nanoparticles and have elicited protective antibody and effective T-helper responses, further supporting the nanoparticle platform as a universal approach for stimulating potent immunogenicity. Here, we present the rational design, structural analysis, and immunogenicity of self-assembling ferritin nanoparticles displaying eight copies of the Neisseria meningitidis trimeric adhesin NadA. We engineered constructs consisting of two different NadA fragments, head only and head with stalk, that we fused to ferritin and expressed in Escherichia coli. Both fusion constructs self-assembled into the expected nanoparticles as determined by Cryo electron microscopy. In mice, the two nanoparticles elicited comparable NadA antibody levels that were 10- to 100-fold higher than those elicited by the corresponding NadA trimer subunits. Further, the NadAferritin nanoparticles potently induced complement-mediated serum bactericidal activity. These findings confirm the value of self-assembling nanoparticles for optimizing the immunogenicity of bacterial antigens and support the broad applicability of the approach to vaccine programs, especially for the presentation of trimeric antigens.


Subject(s)
Nanoparticles , Neisseria meningitidis , Mice , Animals , Ferritins , Antigens, Bacterial , Antigens, Viral , Antibodies, Blocking , Vaccines, Combined , Nanoparticles/chemistry
5.
BMC Pediatr ; 23(1): 76, 2023 02 13.
Article in English | MEDLINE | ID: covidwho-2242649

ABSTRACT

BACKGROUND: Group A streptococcus is human-restricted gram-positive pathogen, responsible for various clinical presentations from mild epidermis infections to life threatened invasive diseases. Under COVID-19 pandemic,. the characteristics of the epidemic strains of GAS could be different. PURPOSE: To investigate epidemiological and molecular features of isolates from GAS infections among children in Beijing, China between January 2020 and December 2021. Antimicrobial susceptibility profiling was performed based on Cinical Laboratory Sandards Institute. Distribution of macrolide-resistance genes, emm types, and superantigens was examined by polymerase chain reaction. RESULTS: 114 GAS isolates were collected which were frequent resistance against erythromycin (94.74%), followed by clindamycin (92.98%), tetracycline (87.72%). Emm12 (46.49%), emm1 (25.44%) were dominant emm types. Distribution of ermB, ermA, and mefA gene was 93.85%, 2.63%, and 14.04%, respectively. Frequent superantigenes identified were smeZ (97.39%), speG (95.65%), and speC (92.17%). Emm1 strains possessed smeZ, ssa, and speC, while emm12 possessed smeZ, ssa, speG, and speC. Erythromycin resistance was predominantly mediated by ermB. Scarlet fever strains harbored smeZ (98.81%), speC (94.05%). Impetigo strains harbored smeZ (88.98%), ssa (88.89%), and speC (88.89%). Psoriasis strains harbored smeZ (100%). CONCLUSIONS: Under COVID-19 pandemic, our collections of GAS infection cutaneous diseases decreased dramatically. Epidemiological analysis of GAS infections among children during COVID-19 pandemic was not significantly different from our previous study. There was a correlation among emm, superantigen gene and disease manifestations. Long-term surveillance and investigation of emm types and superantigens of GAS prevalence are imperative.


Subject(s)
COVID-19 , Streptococcal Infections , Child , Humans , Beijing/epidemiology , Antigens, Bacterial/genetics , COVID-19/epidemiology , Streptococcal Infections/epidemiology , Streptococcus pyogenes , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , China/epidemiology , Erythromycin/pharmacology , Erythromycin/therapeutic use , Superantigens/genetics , Microbial Sensitivity Tests
7.
Int J Mol Sci ; 23(19)2022 Sep 29.
Article in English | MEDLINE | ID: covidwho-2066128

ABSTRACT

COVID-19 patients often develop coagulopathies including microclotting, thrombotic strokes or thrombocytopenia. Autoantibodies are present against blood-related proteins including cardiolipin (CL), serum albumin (SA), platelet factor 4 (PF4), beta 2 glycoprotein 1 (ß2GPI), phosphodiesterases (PDE), and coagulation factors such as Factor II, IX, X and von Willebrand factor (vWF). Different combinations of autoantibodies associate with different coagulopathies. Previous research revealed similarities between proteins with blood clotting functions and SARS-CoV-2 proteins, adenovirus, and bacterial proteins associated with moderate-to-severe COVID-19 infections. This study investigated whether polyclonal antibodies (mainly goat and rabbit) against these viruses and bacteria recognize human blood-related proteins. Antibodies against SARS-CoV-2 and adenovirus recognized vWF, PDE and PF4 and SARS-CoV-2 antibodies also recognized additional antigens. Most bacterial antibodies tested (group A streptococci [GAS], staphylococci, Escherichia coli [E. coli], Klebsiella pneumoniae, Clostridia, and Mycobacterium tuberculosis) cross-reacted with CL and PF4. while GAS antibodies also bound to F2, Factor VIII, Factor IX, and vWF, and E. coli antibodies to PDE. All cross-reactive interactions involved antibody-antigen binding constants smaller than 100 nM. Since most COVID-19 coagulopathy patients display autoantibodies against vWF, PDE and PF4 along with CL, combinations of viral and bacterial infections appear to be necessary to initiate their autoimmune coagulopathies.


Subject(s)
Blood Coagulation Disorders , COVID-19 , Adenoviridae , Animals , Antibodies, Bacterial , Antigens, Bacterial , Autoantibodies , Bacterial Proteins , Blood Coagulation Factors , Capsid Proteins , Cardiolipins , Escherichia coli/metabolism , Factor IX , Factor VIII , Humans , Phosphoric Diester Hydrolases , Platelet Factor 4/metabolism , Prothrombin , Rabbits , SARS-CoV-2 , Serum Albumin , beta 2-Glycoprotein I , von Willebrand Factor
8.
J Transl Med ; 20(1): 389, 2022 09 04.
Article in English | MEDLINE | ID: covidwho-2009423

ABSTRACT

BACKGROUND: Streptococcus pneumoniae is the leading reason for invasive diseases including pneumonia and meningitis, and also secondary infections following viral respiratory diseases such as flu and COVID-19. Currently, serotype-dependent vaccines, which have several insufficiency and limitations, are the only way to prevent pneumococcal infections. Hence, it is plain to need an alternative effective strategy for prevention of this organism. Protein-based vaccine involving conserved pneumococcal protein antigens with different roles in virulence could provide an eligible alternative to existing vaccines. METHODS: In this study, PspC, PhtD and PsaA antigens from pneumococcus were taken to account to predict B-cell and helper T-cell epitopes, and epitope-rich regions were chosen to build the construct. To enhance the immunogenicity of the epitope-based vaccine, a truncated N-terminal fragment of pneumococcal endopeptidase O (PepO) was used as a potential TLR2/4 agonist which was identified by molecular docking studies. The ultimate construct was consisted of the chosen epitope-rich regions, along with the adjuvant role (truncated N-PepO) and suitable linkers. RESULTS: The epitope-based vaccine was assessed as regards physicochemical properties, allergenicity, antigenicity, and toxicity. The 3D structure of the engineered construct was modeled, refined, and validated. Molecular docking and simulation of molecular dynamics (MD) indicated the proper and stable interactions between the vaccine and TLR2/4 throughout the simulation periods. CONCLUSIONS: For the first time this work presents a novel vaccine consisting of epitopes of PspC, PhtD, and PsaA antigens which is adjuvanted with a new truncated domain of PepO. The computational outcomes revealed that the suggested vaccine could be deemed an efficient therapeutic vaccine for S. pneumoniae; nevertheless, in vitro and in vivo examinations should be performed to prove the potency of the candidate vaccine.


Subject(s)
COVID-19 , Streptococcus pneumoniae , Adjuvants, Immunologic , Antigens, Bacterial , Bacterial Proteins , Computational Biology , Epitopes, B-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/chemistry , Humans , Metalloendopeptidases , Molecular Docking Simulation , Molecular Dynamics Simulation , Toll-Like Receptor 2 , Vaccines, Subunit/chemistry
9.
Tuberculosis (Edinb) ; 136: 102253, 2022 09.
Article in English | MEDLINE | ID: covidwho-2004564

ABSTRACT

Tuberculosis (TB) stays a major cause of death globally after COVID-19 and HIV. An early diagnosis to control TB effectively, needs a fast reliable diagnostic method with high sensitivity. Serodiagnosis involving polyclonal antibodies detection against an antigen of Mycobacterium tuberculosis (Mtb) in serum samples can be instrumental. In our study, Rv3874 and Rv3875 antigens were cloned, expressed, and purified individually and as a chimeric construct in Escherichia coli BL21. Enzyme-Linked Immunosorbent Assay (ELISA) based findings revealed that the Rv3874-Rv3875 chimeric construct was two-fold more sensitive (59.7%) than the individual sensitivities of Rv3874 (28.4%) and Rv3875 (24.9%) for 201 serum TB positive samples. Furthermore, the fusion construct was a little more sensitive (60.4%) for male subjects than that for females (58.8%). Lastly, our preliminary findings, molecular insights of secondary structure, and statistical and in silico analysis of each construct also advocate that CEP can be considered a better immunodiagnostic tool in addition to previously reported EC skin test.


Subject(s)
COVID-19 , Mycobacterium tuberculosis , Tuberculosis , Antigens, Bacterial , Enzyme-Linked Immunosorbent Assay/methods , Escherichia coli , Female , Humans , Male , Mycobacterium tuberculosis/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sensitivity and Specificity , Serologic Tests , Tuberculosis/diagnosis
10.
Tuberculosis (Edinb) ; 134: 102186, 2022 05.
Article in English | MEDLINE | ID: covidwho-1783793

ABSTRACT

COVID-19 has affected the progress made in the prevention and treatment of tuberculosis (TB); hence, the mortality of tuberculosis has risen. Different strategies-based novel TB vaccine candidates have been developed. This study identifies strategies to overcome the limitations of Bacille Calmette-Guérin (BCG) in preventing latent infection and reactivation of TB. The latency antigen Rv0572c was selected based on the mechanism of interaction between Mycobacterium tuberculosis and its host. The rRv0572c protein was used to stimulate whole blood samples derived from patients with clinically diagnosed active TB (ATBs) or latent TB infections (LTBIs) and healthy control (HCs) donors, confirming that this protein can be recognized by T cells in patients with TB, especially LTBIs. C57BL/6 mice were used to investigate the immunogenicity of the rRv0572c protein emulsified in the liposome adjuvant dimethyldioctadecylammonium [DDA], monophosphoryl lipid A [MPLA], trehalose-6, 6'-dibehenate [TDB] (DMT). The results demonstrated that rRv0572c/DMT could boost BCG-primed mice to induce antigen-specific CD4+ T cell production and generate functional T cells dominated by antigen-specific CD8+ T cells. The rRv0572c/DMT vaccine could also trigger limited Th2 humoral immune responses. These findings suggest that rRv0572c/DMT is a potential subunit vaccine candidate that can be used as a booster vaccine for BCG.


Subject(s)
COVID-19 , Latent Tuberculosis , Mycobacterium tuberculosis , Tuberculosis Vaccines , Tuberculosis , Adjuvants, Immunologic , Animals , Antigens, Bacterial , BCG Vaccine , CD8-Positive T-Lymphocytes , Humans , Liposomes , Mice , Mice, Inbred C57BL , Tuberculosis/prevention & control , Vaccines, Subunit
11.
EBioMedicine ; 78: 103939, 2022 Apr.
Article in English | MEDLINE | ID: covidwho-1763702

ABSTRACT

Rapid, accurate, sputum-free tests for tuberculosis (TB) triage and confirmation are urgently needed to close the widening diagnostic gap. We summarise key technologies and review programmatic, systems, and resource issues that could affect the impact of diagnostics. Mid-to-early-stage technologies like artificial intelligence-based automated digital chest X-radiography and capillary blood point-of-care assays are particularly promising. Pitfalls in the diagnostic pipeline, included a lack of community-based tools. We outline how these technologies may complement one another within the context of the TB care cascade, help overturn current paradigms (eg, reducing syndromic triage reliance, permitting subclinical TB to be diagnosed), and expand options for extra-pulmonary TB. We review challenges such as the difficulty of detecting paucibacillary TB and the limitations of current reference standards, and discuss how researchers and developers can better design and evaluate assays to optimise programmatic uptake. Finally, we outline how leveraging the urgency and innovation applied to COVID-19 is critical to improving TB patients' diagnostic quality-of-care.


Subject(s)
COVID-19 , Mycobacterium tuberculosis , Tuberculosis , Antigens, Bacterial , Artificial Intelligence , COVID-19/diagnosis , Humans , Sputum , Tuberculosis/diagnosis
12.
Int J Mol Sci ; 23(5)2022 Feb 28.
Article in English | MEDLINE | ID: covidwho-1736946

ABSTRACT

Mycobacterium tuberculosis (Mtb) represents a major burden to global health, and refined vaccines are needed. Replication-deficient lymphocytic choriomeningitis virus (rLCMV)-based vaccine vectors against cytomegalovirus have proven safe for human use and elicited robust T cell responses in a large proportion of vaccine recipients. Here, we developed an rLCMV vaccine expressing the Mtb antigens TB10.4 and Ag85B. In mice, rLCMV elicited high frequencies of polyfunctional Mtb-specific CD8 and CD4 T cell responses. CD8 but not CD4 T cells were efficiently boosted upon vector re-vaccination. High-frequency responses were also observed in neonatally vaccinated mice, and co-administration of rLCMV with Expanded Program of Immunization (EPI) vaccines did not result in substantial reciprocal interference. Importantly, rLCMV immunization significantly reduced the lung Mtb burden upon aerosol challenge, resulting in improved lung ventilation. Protection was associated with increased CD8 T cell recruitment but reduced CD4 T cell infiltration upon Mtb challenge. When combining rLCMV with BCG vaccination in a heterologous prime-boost regimen, responses to the rLCMV-encoded Mtb antigens were further augmented, but protection was not significantly different from rLCMV or BCG vaccination alone. This work suggests that rLCMV may show utility for neonatal and/or adult vaccination efforts against pulmonary tuberculosis.


Subject(s)
Mycobacterium tuberculosis , Animals , Antigens, Bacterial , BCG Vaccine , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Lymphocytic choriomeningitis virus/genetics , Mice , Mycobacterium tuberculosis/genetics
13.
Int J Infect Dis ; 113 Suppl 1: S82-S87, 2021 Dec.
Article in English | MEDLINE | ID: covidwho-1575296

ABSTRACT

OBJECTIVES: The interaction of COVID-19 and tuberculosis (TB) are still poor characterized. Here we evaluated the immune response specific for Micobacterium tuberculosis (Mtb) and SARS-CoV-2 using a whole-blood-based assay-platform in COVID-19 patients either with TB or latent TB infection (LTBI). METHODS: We evaluated IFN-γ level in plasma from whole-blood stimulated with Mtb antigens in the Quantiferon-Plus format or with peptides derived from SARS-CoV-2 spike protein, Wuhan-Hu-1 isolate (CD4-S). RESULTS: We consecutively enrolled 63 COVID-19, 10 TB-COVID-19 and 11 LTBI-COVID-19 patients. IFN-γ response to Mtb-antigens was significantly associated to TB status and therefore it was higher in TB-COVID-19 and LTBI-COVID-19 patients compared to COVID-19 patients (p ≤ 0.0007). Positive responses against CD4-S were found in 35/63 COVID-19 patients, 7/11 LTBI-COVID-19 and only 2/10 TB-COVID-19 patients. Interestingly, the responders in the TB-COVID-19 group were less compared to COVID-19 and LTBI-COVID-19 groups (p = 0.037 and 0.044, respectively). Moreover, TB-COVID-19 patients showed the lowest quantitative IFN-γ response to CD4-S compared to COVID-19-patients (p = 0.0336) and LTBI-COVID-19 patients (p = 0.0178). CONCLUSIONS: Our data demonstrate that COVID-19 patients either TB or LTBI have a low ability to build an immune response to SARS-CoV-2 while retaining the ability to respond to Mtb-specific antigens.


Subject(s)
COVID-19 , Coinfection , Tuberculosis , Antigens, Bacterial/immunology , Antigens, Viral/immunology , COVID-19/immunology , Humans , Interferon-gamma/immunology , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Tuberculosis/immunology
14.
Cytokine ; 150: 155785, 2022 02.
Article in English | MEDLINE | ID: covidwho-1568622

ABSTRACT

SARS-CoV-2 and latent Mycobacterium tuberculosis infection are both highly co-prevalent in many parts of the globe. Whether exposure to SARS-CoV-2 influences the antigen specific immune responses in latent tuberculosis has not been investigated. We examined the baseline, mycobacterial antigen and mitogen induced cytokine and chemokine responses in latent tuberculosis (LTBI) individuals with or without SARS-CoV-2 seropositivity, LTBI negative individuals with SARS-CoV-2 seropositivity and healthy control (both LTBI and SARS-CoV-2 negative) individuals. Our results demonstrated that LTBI individuals with SARS-CoV-2 seropositivity (LTBI+/IgG +) were associated with increased levels of unstimulated and TB-antigen stimulated IFNγ, IL-2, TNFα, IL-17, IL-1ß, IL-6, IL-12, IL-4, CXCL1, CXCL9 and CXCL10 when compared to those without seropositivity (LTBI+/IgG-). In contrast, LTBI+/IgG+ individuals were associated with decreased levels of IL-5 and IL-10. No significant difference in the levels of cytokines/chemokines was observed upon mitogen stimulation between the groups. SARS-CoV-2 seropositivity was associated with enhanced unstimulated and TB-antigen stimulated but not mitogen stimulated production of cytokines and chemokines in LTBI+ compared to LTBI negative individuals. Finally, most of these significant differences were not observed when LTBI negative individuals with SARS-CoV-2 seropositivity and controls were examined. Our data clearly demonstrate that both baseline and TB - antigen induced cytokine responses are augmented in the presence of SARS-CoV-2 seropositivity, suggesting an augmenting effect of prior SARS-CoV-2 infection on the immune responses of LTBI individuals.


Subject(s)
COVID-19/complications , Cytokines/blood , Latent Tuberculosis/complications , SARS-CoV-2/immunology , Aged , Aged, 80 and over , Antibodies, Viral/blood , Antigens, Bacterial/immunology , COVID-19/immunology , Chemokines/blood , Female , Humans , Immunocompromised Host , Immunoglobulin G/blood , Inflammation , Latent Tuberculosis/blood , Latent Tuberculosis/immunology , Lymphocyte Activation/drug effects , Male , Middle Aged , Phytohemagglutinins/pharmacology , Seroconversion
15.
Sci Rep ; 11(1): 19193, 2021 09 28.
Article in English | MEDLINE | ID: covidwho-1442802

ABSTRACT

Outside of the ongoing COVID-19 pandemic, tuberculosis is the leading cause of infectious disease mortality globally. Currently, there is no commercially available point-of-care diagnostic that is rapid, inexpensive, and highly sensitive for the diagnosis of active tuberculosis disease. Here we describe the development and optimization of a novel, highly sensitive prototype bioelectronic tuberculosis antigen (BETA) assay to detect tuberculosis-specific antigen, CFP10, in small-volume serum and urine samples. In this proof-of-concept study we evaluated the performance of the BETA assay using clinical specimens collected from presumptive tuberculosis patients from three independent cohorts. Circulating CFP10 antigen was detected in ALL serum (n = 19) and urine (n = 3) samples from bacteriologically confirmed tuberculosis patients who were untreated or had less than one week of treatment at time of serum collection, successfully identifying all culture positive tuberculosis patients. No CFP10 antigen was detected in serum (n = 7) or urine (n = 6) samples from individuals who were determined to be negative for tuberculosis disease. Additionally, antigen quantification using the BETA assay of paired serum samples collected from tuberculosis patients (n = 8) both before and after treatment initiation, indicate consistently declining within-person levels of CFP10 antigen during treatment. This novel, low-cost assay demonstrates potential as a rapid, non-sputum-based, point-of-care tool for the diagnosis of tuberculosis disease.


Subject(s)
Diagnostic Tests, Routine/methods , Peptide Fragments , Tuberculosis/diagnosis , Antigens, Bacterial/blood , Antigens, Bacterial/isolation & purification , Antigens, Bacterial/urine , Mycobacterium tuberculosis/immunology , Peptide Fragments/blood , Peptide Fragments/isolation & purification , Peptide Fragments/urine , Sensitivity and Specificity , Tuberculosis, Pulmonary/diagnosis
16.
J Clin Monit Comput ; 36(5): 1289-1295, 2022 10.
Article in English | MEDLINE | ID: covidwho-1437302

ABSTRACT

The COVID-19 pandemic has caused personal protective equipment shortages worldwide and required healthcare workers to develop novel ways of protecting themselves. Anesthesiologists in particular are exposed to increased risks of contamination when performing interventions such as airway manipulations. We developed and tested an aerosolization protective device which contains aerosols around the patient's airway and helps eliminate particles using negative pressure. This intubation box is a polymethyl methacrylate prism with openings for gloves, integrated suction and ventilation connectors. We conducted a randomised controlled series of tests to detect 0.5 µm particles after a simulated cough inside the intubation box, using a high-fidelity simulation mannequin. Setting and main outcome: We measured particle concentrations inside the box with and without suction turned on, in both negative and positive pressure operating rooms. We also obtained particle concentrations outside our box and compared them to non-airtight barrier devices. One minute following simulated cough, the mean number of particles per cubic foot in our box with suction on is around 45% that with the suction off (1,462,373 vs 3,272,080, P < 0.0001) in the negative pressure room, and four times lower than with the suction off (760,380 vs 3,088,700, P < 0.0001) in the positive pressure room. After a simulated cough inside the box, particles can be detected in front of the anesthesiologist's face with a non-airtight device, while none are detected when our box is sealed and its suction turned on. The use of our negative pressure intubation box prevents contamination of surroundings and increases particle elimination, regardless of room pressure.


Subject(s)
COVID-19 , Infectious Disease Transmission, Patient-to-Professional , Antigens, Bacterial , Cough , Humans , Infectious Disease Transmission, Patient-to-Professional/prevention & control , Intubation, Intratracheal , Pandemics/prevention & control , Polymethyl Methacrylate , Respiratory Aerosols and Droplets , Vacuum
17.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Article in English | MEDLINE | ID: covidwho-1387607

ABSTRACT

The global incidence of tuberculosis remains unacceptably high, with new preventative strategies needed to reduce the burden of disease. We describe here a method for the generation of synthetic self-adjuvanted protein vaccines and demonstrate application in vaccination against Mycobacterium tuberculosis Two vaccine constructs were designed, consisting of full-length ESAT6 protein fused to the TLR2-targeting adjuvants Pam2Cys-SK4 or Pam3Cys-SK4 These were produced by chemical synthesis using a peptide ligation strategy. The synthetic self-adjuvanting vaccines generated powerful local CD4+ T cell responses against ESAT6 and provided significant protection in the lungs from virulent M. tuberculosis aerosol challenge when administered to the pulmonary mucosa of mice. The flexible synthetic platform we describe, which allows incorporation of adjuvants to multiantigenic vaccines, represents a general approach that can be applied to rapidly assess vaccination strategies in preclinical models for a range of diseases, including against novel pandemic pathogens such as SARS-CoV-2.


Subject(s)
Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/pharmacology , Tuberculosis/immunology , Tuberculosis/prevention & control , Vaccines, Conjugate/pharmacology , Adjuvants, Immunologic/pharmacology , Animals , Antigens, Bacterial/immunology , BCG Vaccine/immunology , BCG Vaccine/pharmacology , Bacterial Proteins , CD4-Positive T-Lymphocytes/immunology , COVID-19/prevention & control , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , SARS-CoV-2/immunology , Toll-Like Receptor 2/immunology , Tuberculosis Vaccines/immunology , Vaccines, Conjugate/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/pharmacology
18.
Int J Mol Sci ; 22(8)2021 Apr 08.
Article in English | MEDLINE | ID: covidwho-1299441

ABSTRACT

Pneumonia due to respiratory infection with most prominently bacteria, but also viruses, fungi, or parasites is the leading cause of death worldwide among all infectious disease in both adults and infants. The introduction of modern antibiotic treatment regimens and vaccine strategies has helped to lower the burden of bacterial pneumonia, yet due to the unavailability or refusal of vaccines and antimicrobials in parts of the global population, the rise of multidrug resistant pathogens, and high fatality rates even in patients treated with appropriate antibiotics pneumonia remains a global threat. As such, a better understanding of pathogen virulence on the one, and the development of innovative vaccine strategies on the other hand are once again in dire need in the perennial fight of men against microbes. Recent data show that the secretome of bacteria consists not only of soluble mediators of virulence but also to a significant proportion of extracellular vesicles-lipid bilayer-delimited particles that form integral mediators of intercellular communication. Extracellular vesicles are released from cells of all kinds of organisms, including both Gram-negative and Gram-positive bacteria in which case they are commonly termed outer membrane vesicles (OMVs) and membrane vesicles (MVs), respectively. (O)MVs can trigger inflammatory responses to specific pathogens including S. pneumonia, P. aeruginosa, and L. pneumophila and as such, mediate bacterial virulence in pneumonia by challenging the host respiratory epithelium and cellular and humoral immunity. In parallel, however, (O)MVs have recently emerged as auspicious vaccine candidates due to their natural antigenicity and favorable biochemical properties. First studies highlight the efficacy of such vaccines in animal models exposed to (O)MVs from B. pertussis, S. pneumoniae, A. baumannii, and K. pneumoniae. An advanced and balanced recognition of both the detrimental effects of (O)MVs and their immunogenic potential could pave the way to novel treatment strategies in pneumonia and effective preventive approaches.


Subject(s)
Bacteria/metabolism , Bacterial Outer Membrane/metabolism , Extracellular Vesicles/metabolism , Pneumonia, Bacterial/microbiology , Adaptive Immunity , Animals , Antigens, Bacterial/immunology , Bacteria/immunology , Bacterial Outer Membrane/immunology , Bacterial Vaccines/immunology , Host-Pathogen Interactions/immunology , Humans , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/prevention & control , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/prevention & control , Virulence
19.
Trends Microbiol ; 29(11): 1034-1045, 2021 11.
Article in English | MEDLINE | ID: covidwho-1240633

ABSTRACT

Antimicrobial resistance is an increasing global threat and alternative treatments substituting failing antibiotics are urgently needed. Vaccines are recognized as highly effective tools to mitigate antimicrobial resistance; however, the selection of bacterial antigens as vaccine candidates remains challenging. In recent years, advances in mass spectrometry-based proteomics have led to the development of so-called immunopeptidomics approaches that allow the untargeted discovery of bacterial epitopes that are presented on the surface of infected cells. Especially for intracellular bacterial pathogens, immunopeptidomics holds great promise to uncover antigens that can be encoded in viral vector- or nucleic acid-based vaccines. This review provides an overview of immunopeptidomics studies on intracellular bacterial pathogens and considers future directions and challenges in advancing towards next-generation vaccines.


Subject(s)
Bacterial Vaccines , Vaccine Development , Antigens, Bacterial , Mass Spectrometry , Proteomics
20.
mBio ; 12(2)2021 04 20.
Article in English | MEDLINE | ID: covidwho-1195824

ABSTRACT

New vaccines are urgently needed against Mycobacterium tuberculosis (Mtb), which kills more than 1.4 million people each year. CD4 T cell differentiation is a key determinant of protective immunity against Mtb, but it is not fully understood how host-pathogen interactions shape individual antigen-specific T cell populations and their protective capacity. Here, we investigated the immunodominant Mtb antigen, MPT70, which is upregulated in response to gamma interferon (IFN-γ) or nutrient/oxygen deprivation of in vitro-infected macrophages. Using a murine aerosol infection model, we compared the in vivo expression kinetics of MPT70 to a constitutively expressed antigen, ESAT-6, and analyzed their corresponding CD4 T cell phenotype and vaccine protection. For wild-type Mtb, we found that in vivo expression of MPT70 was delayed compared to ESAT-6. This delayed expression was associated with induction of less differentiated MPT70-specific CD4 T cells but, compared to ESAT-6, also reduced protection after vaccination. In contrast, infection with an MPT70-overexpressing Mtb strain promoted highly differentiated KLRG1+CX3CR1+ CD4 T cells with limited lung-homing capacity. Importantly, this differentiated phenotype could be prevented by vaccination, and against the overexpressing strain, vaccination with MPT70 conferred protection similar to vaccination with ESAT-6. Together, our data indicate that high in vivo antigen expression drives T cells toward terminal differentiation and that targeted vaccination with adjuvanted protein can counteract this phenomenon by maintaining T cells in a protective less differentiated state. These observations shed new light on host-pathogen interactions and provide guidance on how future Mtb vaccines can be designed to tip the immune balance in favor of the host.IMPORTANCE Tuberculosis, caused by Mtb, constitutes a global health crisis of massive proportions and the impact of the current coronavirus disease 2019 (COVID-19) pandemic is expected to cause a rise in tuberculosis-related deaths. Improved vaccines are therefore needed more than ever, but a lack of knowledge on protective immunity hampers their development. The present study shows that constitutively expressed antigens with high availability drive highly differentiated CD4 T cells with diminished protective capacity, which could be a survival strategy by Mtb to evade T cell immunity against key antigens. We demonstrate that immunization with such antigens can counteract this phenomenon by maintaining antigen-specific T cells in a state of low differentiation. Future vaccine strategies should therefore explore combinations of multiple highly expressed antigens and we suggest that T cell differentiation could be used as a readily measurable parameter to identify these in both preclinical and clinical studies.


Subject(s)
Antigens, Bacterial/immunology , CD4-Positive T-Lymphocytes/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/pharmacology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/prevention & control , Animals , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Bacterial Proteins/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/microbiology , Cell Differentiation/immunology , Disease Models, Animal , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Gene Expression , Genes, Bacterial , Humans , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mycobacterium tuberculosis/genetics , Tuberculosis Vaccines/genetics , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL