Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 117.383
Filtrer
1.
PLoS One ; 19(7): e0305137, 2024.
Article de Anglais | MEDLINE | ID: mdl-38950036

RÉSUMÉ

Electrospun (ES) fibrous nanomaterials have been widely investigated as novel biomaterials. These biomaterials have to be safe and biocompatible; hence, they need to be tested for cytotoxicity before being administered to patients. The aim of this study was to develop a suitable and biorelevant in vitro cytotoxicity assay for ES biomaterials (e.g. wound dressings). We compared different in vitro cytotoxicity assays, and our model wound dressing was made from polycaprolactone and polyethylene oxide and contained chloramphenicol as the active pharmaceutical ingredient. Baby Hamster Kidney cells (BHK-21), human primary fibroblasts and MTS assays together with real-time cell analysis were selected. The extract exposure and direct contact safety evaluation setups were tested together with microscopic techniques. We found that while extract exposure assays are suitable for the initial testing, the biocompatibility of the biomaterial is revealed in in vitro direct contact assays where cell interactions with the ES wound dressing are evaluated. We observed significant differences in the experimental outcome, caused by the experimental set up modification such as cell line choice, cell medium and controls used, conducting the phosphate buffer washing step or not. A more detailed technical protocol for the in vitro cytotoxicity assessment of ES wound dressings was developed.


Sujet(s)
Bandages , Matériaux biocompatibles , Cicatrisation de plaie , Animaux , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Humains , Lignée cellulaire , Test de matériaux , Cricetinae , Polyesters/composition chimique , Fibroblastes/effets des médicaments et des substances chimiques , Anti-infectieux/pharmacologie , Polyéthylène glycols/composition chimique , Chloramphénicol/pharmacologie
2.
Vopr Virusol ; 69(3): 219-230, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38996371

RÉSUMÉ

INTRODUCTION: Specific prevention of a number of infectious diseases has been introduced into the vaccination schedule. The production of immunoprophylactic drugs, in order to establish standard properties, including safety and specific effectiveness, requires strict adherence to manufacturing regulations, and the reliability of the results obtained requires monitoring of these parameters. The specific effectiveness of vaccine preparations is standardized according to the indicators of stimulation of specific antibody response formed in the body of vaccinated model biological objects. OBJECTIVE: Determination of the immune reactivity of white mice to vaccination with the QazVac vaccine to establish the possibility of using them as a biological model in assessing the immunogenicity of the vaccine instead of Syrian hamsters. MATERIALS AND METHODS: The immune reactivity of model animals was assessed by the seroconversion rate, dynamics of antibody titers to the SARS-CoV-2 virus formed in the body after vaccination with the test vaccine. In the case of seropositivity of animals before administration of vaccine or placebo, the level of immune reactivity was calculated by the difference in antibody titers between control and vaccinated animals or by the difference in antibody titers before and after immunization. Specific antibodies were detected and their titer was determined using a neutralization reaction. RESULTS: The research results showed that the tested biological models had approximately the same immune reactivity to the administration of the QazVac vaccine, confirmed by the level and dynamics of antibody titers. When analyzing the fold increase in antibody titers in comparison to those of control animals, Syrian hamsters were more reactive compared to mice. But SPF white mice were standardized in their lack of the immune reactivity to SARS-CoV-2 virus before the immunization. CONCLUSION: The data obtained indicate that the immune reactivity of white mice to the administration of the QazVac vaccine in terms of the rate and dynamics of the formation of virus-neutralizing antibodies is approximately equivalent to the immune reactivity of Syrian hamsters. Before immunization with the vaccine, SPF white mice, in contrast to Syrian hamsters, do not have humoral immunity specific to the SARS-CoV-2 virus. The immune reactivity equivalent to that observed of Syrian hamsters and the absence of antibodies to the SARS-CoV-2 virus at a baseline indicate the superiority of the use of white mice in assessing the immunogenicity of vaccines against COVID-19 and/or obtaining specific factors of humoral immunity.


Sujet(s)
Anticorps antiviraux , Vaccins contre la COVID-19 , COVID-19 , SARS-CoV-2 , Vaccination , Vaccins inactivés , Animaux , COVID-19/prévention et contrôle , COVID-19/immunologie , SARS-CoV-2/immunologie , Souris , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/administration et posologie , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Vaccins inactivés/immunologie , Vaccins inactivés/administration et posologie , Cricetinae , Mesocricetus , Immunogénicité des vaccins , Humains , Modèles animaux de maladie humaine , Immunité humorale , Femelle , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie
3.
Front Immunol ; 15: 1408510, 2024.
Article de Anglais | MEDLINE | ID: mdl-39021566

RÉSUMÉ

Equid alphaherpesvirus 1 (EqAHV1) is a viral pathogen known to cause respiratory disease, neurologic syndromes, and abortion storms in horses. Currently, there are no vaccines that provide complete protection against EqAHV1. Marker vaccines and the differentiation of infected and vaccinated animals (DIVA) strategy are effective for preventing and controlling outbreaks but have not been used for the prevention of EqAHV1 infection. Glycoprotein 2 (gp2), located on the envelope of viruses (EqAHV1), exhibits high antigenicity and functions as a molecular marker for DIVA. In this study, a series of EqAHV1 mutants with deletion of gp2 along with other virulence genes (TK, UL24/TK, gI/gE) were engineered. The mutant viruses were studied in vitro and then in an in vivo experiment using Golden Syrian hamsters to assess the extent of viral attenuation and the immune response elicited by the mutant viruses in comparison to the wild-type (WT) virus. Compared with the WT strain, the YM2019 Δgp2, ΔTK/gp2, and ΔUL24/TK/gp2 strains exhibited reduced growth in RK-13 cells, while the ΔgI/gE/gp2 strain exhibited significantly impaired proliferation. The YM2019 Δgp2 strain induced clinical signs and mortality in hamsters. In contrast, the YM2019 ΔTK/gp2 and ΔUL24/TK/gp2 variants displayed diminished pathogenicity, causing no observable clinical signs or fatalities. Immunization with nasal vaccines containing YM2019 ΔTK/gp2 and ΔUL24/TK/gp2 elicited a robust immune response in hamsters. In particular, compared with the vaccine containing the ΔTK/gp2 strain, the vaccine containing the ΔUL24/TK/gp2 strain demonstrated enhanced immune protection upon challenge with the WT virus. Furthermore, an ELISA for gp2 was established and refined to accurately differentiate between infected and vaccinated animals. These results confirm that the ΔUL24/TK/gp2 strain is a safe and effective live attenuated vaccine candidate for controlling EqAHV1 infection.


Sujet(s)
Infections à Herpesviridae , Herpèsvirus équin de type 1 , Vaccins atténués , Animaux , Vaccins atténués/immunologie , Infections à Herpesviridae/prévention et contrôle , Infections à Herpesviridae/immunologie , Infections à Herpesviridae/virologie , Infections à Herpesviridae/médecine vétérinaire , Herpèsvirus équin de type 1/immunologie , Herpèsvirus équin de type 1/génétique , Equus caballus , Mesocricetus , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/génétique , Cricetinae , Maladies des chevaux/prévention et contrôle , Maladies des chevaux/immunologie , Maladies des chevaux/virologie , Vaccins antiviraux/immunologie , Vaccins antiviraux/génétique , Lignée cellulaire , Mutation
4.
J Med Microbiol ; 73(7)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39028257

RÉSUMÉ

Clostridioides difficile is the most common cause of nosocomial antibiotic-associated diarrhoea and is responsible for a spectrum of diseases characterized by high levels of recurrence and morbidity. In some cases, complications can lead to death. Currently, several types of animal models have been developed to study various aspects of C. difficile infection (CDI), such as colonization, virulence, transmission and recurrence. These models have also been used to test the role of environmental conditions, such as diet, age and microbiome that modulate infection outcome, and to evaluate several therapeutic strategies. Different rodent models have been used successfully, such as the hamster model and the gnotobiotic and conventional mouse models. These models can be applied to study either the initial CDI infectious process or recurrences. The applications of existing rodent models and their advantages and disadvantages are discussed here.


Sujet(s)
Clostridioides difficile , Infections à Clostridium , Modèles animaux de maladie humaine , Animaux , Infections à Clostridium/microbiologie , Clostridioides difficile/pathogénicité , Souris , Cricetinae , Humains , Rodentia/microbiologie , Axénie
5.
Biotechnol J ; 19(6): e2400251, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-39031790

RÉSUMÉ

The Chinese hamster ovary (CHO) cell is an epithelial-like cell that produces proteins with post-translational modifications similar to human glycosylation. It is widely used in the production of recombinant therapeutic proteins and monoclonal antibodies. Culturing CHO cells typically requires the addition of a certain proportion of fetal bovine serum (FBS) to maintain cell proliferation and passaging. However, serum is characterized by its complex composition, batch-to-batch variability, high cost, and potential risk of exogenous contaminants such as mycoplasma and viruses, which impact the purity and safety of the synthesized proteins. Therefore, search for serum alternatives and development of serum-free media for CHO-based protein biomanufacturing are of great significance. This review systematically summarizes the application advantages of CHO cells and strategies for high-density expression. It highlights the developmental trends of serum substitutes from human platelet lysates to animal-free extracts and microbial-derived substances and elucidates the mechanisms by which these substitutes enhance CHO cell culture performance and recombinant protein production, aiming to provide theoretical guidance for exploring novel serum alternatives and developing serum-free media for CHO cells.


Sujet(s)
Cricetulus , Protéines recombinantes , Cellules CHO , Animaux , Milieux de culture sans sérum , Protéines recombinantes/métabolisme , Humains , Techniques de culture cellulaire/méthodes , Cricetinae , Prolifération cellulaire
6.
Zhonghua Yu Fang Yi Xue Za Zhi ; 58(7): 959-966, 2024 Jul 06.
Article de Chinois | MEDLINE | ID: mdl-39034780

RÉSUMÉ

Objective: To establish a Plaque-reduction Neutralization Test (PRNT) for the detection of neutralizing antibody titers of Human Respiratory Syncytial Virus (HRSV) and optimize the conditions for preliminary application. Methods: The CHO expression system was used to produce palivizumab monoclonal antibody (palivizumab) and the influencing factors such as cell type, cell culture duration, fixation and permeabilization protocols, and blocking agents. The reproducibility of the method was verified and its correlation was verified with conventional PRNT. Finally, the optimized PRNT assay was further used to determine neutralizing antibody titers against HRSV subtypes A and B in BALB/c mouse serum (immunized by intramuscular injection of HRSV fusion proteins). Results: Palivizumab was expressed at approximately 50 mg/L. The optimal working conditions for PRNT were as follows: culturing HEp-2 cells for 2 days, fixing with 4% (V/V) paraformaldehyde at room temperature for 15 min followed by 0.2% (V/V) Triton X-100 permeabilization for 15 minutes as the optimal fixation-permeabilization and removing the blocking step. The overall coefficient of variation (CV) for the reproducibility validation of this method was <15%, showing a good linear relationship with the conventional PRNT. The Spearman correlation coefficient rs was 0.983. This method was used to detect neutralizing antibody titers in mouse sera against HRSV subtype A strain long and subtype B strain 9320, and the fusion proteins combined with AlOH and CpG adjuvant induced the highest neutralizing antibody titers in mice. Conclusion: The HRSV neutralizing antibody assay established in this study is rapid, reproducible, high-throughput, and can be used to detect neutralizing antibodies to HRSV subtypes A and B.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Souris de lignée BALB C , Tests de neutralisation , Virus respiratoire syncytial humain , Animaux , Anticorps neutralisants/immunologie , Souris , Virus respiratoire syncytial humain/immunologie , Humains , Tests de neutralisation/méthodes , Anticorps antiviraux/immunologie , Cellules CHO , Cricetulus , Cricetinae , Palivizumab
7.
PLoS Pathog ; 20(7): e1012345, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38968329

RÉSUMÉ

The CRISPR-Cas13 system has been proposed as an alternative treatment of viral infections. However, for this approach to be adopted as an antiviral, it must be optimized until levels of efficacy rival or exceed the performance of conventional approaches. To take steps toward this goal, we evaluated the influenza viral RNA degradation patterns resulting from the binding and enzymatic activity of mRNA-encoded LbuCas13a and two crRNAs from a prior study, targeting PB2 genomic and messenger RNA. We found that the genome targeting guide has the potential for significantly higher potency than originally detected, because degradation of the genomic RNA is not uniform across the PB2 segment, but it is augmented in proximity to the Cas13 binding site. The PB2 genome targeting guide exhibited high levels (>1 log) of RNA degradation when delivered 24 hours post-infection in vitro and maintained that level of degradation over time, with increasing multiplicity of infection (MOI), and across modern influenza H1N1 and H3N2 strains. Chemical modifications to guides with potent LbuCas13a function, resulted in nebulizer delivered efficacy (>1-2 log reduction in viral titer) in a hamster model of influenza (Influenza A/H1N1/California/04/09) infection given prophylactically or as a treatment (post-infection). Maximum efficacy was achieved with two doses, when administered both pre- and post-infection. This work provides evidence that mRNA-encoded Cas13a can effectively mitigate Influenza A infections opening the door to the development of a programmable approach to treating multiple respiratory infections.


Sujet(s)
Systèmes CRISPR-Cas , Grippe humaine , Stabilité de l'ARN , ARN messager , ARN viral , Animaux , ARN viral/génétique , ARN viral/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Humains , Grippe humaine/virologie , Sous-type H1N1 du virus de la grippe A/génétique , Sous-type H3N2 du virus de la grippe A/génétique , Infections à Orthomyxoviridae/virologie , Antiviraux/pharmacologie , Chiens , Cricetinae , Protéines virales/génétique , Protéines virales/métabolisme , Mesocricetus , Cellules rénales canines Madin-Darby
8.
Biophys Chem ; 312: 107287, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38981174

RÉSUMÉ

Although intracellular ultrastructures have typically been studied using microscopic techniques, it is difficult to observe ultrastructures at the submicron scale of living cells due to spatial resolution (fluorescence microscopy) or high vacuum environment (electron microscopy). We investigate the nanometer scale intracellular ultrastructures of living CHO cells in various osmolality using small-angle X-ray scattering (SAXS), and especially the structures of ribosomes, DNA double helix, and plasma membranes in-cell environment are observed. Ribosomes expand and contract in response to osmotic pressure, and the inter-ribosomal correlation occurs under isotonic and hyperosmolality. The DNA double helix is not dependent on the osmotic pressure. Under high osmotic pressure, the plasma membrane folds into form a multilamellar structure with a periodic length of about 6 nm. We also study the ultrastructural changes caused by formaldehyde fixation, freezing and heating.


Sujet(s)
Membrane cellulaire , Cricetulus , Pression osmotique , Diffusion aux petits angles , Diffraction des rayons X , Animaux , Cellules CHO , Cricetinae , Membrane cellulaire/composition chimique , ADN/composition chimique , Ribosomes/composition chimique , Ribosomes/métabolisme , Formaldéhyde/composition chimique , Congélation
9.
Nat Commun ; 15(1): 6053, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39025863

RÉSUMÉ

Respiratory viral infections cause morbidity and mortality worldwide. Despite the success of vaccines, vaccination efficacy is weakened by the rapid emergence of viral variants with immunoevasive properties. The development of an off-the-shelf, effective, and safe therapy against respiratory viral infections is thus desirable. Here, we develop NanoSTING, a nanoparticle formulation of the endogenous STING agonist, 2'-3' cGAMP, to function as an immune activator and demonstrate its safety in mice and rats. A single intranasal dose of NanoSTING protects against pathogenic strains of SARS-CoV-2 (alpha and delta VOC) in hamsters. In transmission experiments, NanoSTING reduces the transmission of SARS-CoV-2 Omicron VOC to naïve hamsters. NanoSTING also protects against oseltamivir-sensitive and oseltamivir-resistant strains of influenza in mice. Mechanistically, NanoSTING upregulates locoregional interferon-dependent and interferon-independent pathways in mice, hamsters, as well as non-human primates. Our results thus implicate NanoSTING as a broad-spectrum immune activator for controlling respiratory virus infection.


Sujet(s)
Administration par voie nasale , Nanoparticules , SARS-CoV-2 , Animaux , Nanoparticules/composition chimique , Nanoparticules/administration et posologie , Souris , Cricetinae , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/immunologie , Modèles animaux de maladie humaine , Humains , Protéines membranaires/agonistes , Protéines membranaires/métabolisme , Femelle , Nucléotides cycliques/pharmacologie , Rats , COVID-19/prévention et contrôle , COVID-19/immunologie , COVID-19/virologie , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/virologie , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/traitement médicamenteux , Mâle , Antiviraux/pharmacologie , Antiviraux/administration et posologie , Souris de lignée C57BL
10.
J Med Virol ; 96(7): e29782, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39011762

RÉSUMÉ

Extracellular vesicles (EVs) are shown to be a novel viral transmission model capable of increasing a virus's tropism. According to our earlier research, cells infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or transfected with envelope protein plasmids generate a novel type of EVs that are micrometer-sized and able to encase virus particles. Here, we showed the capacity of these EVs to invade various animals both in vitro and in vivo independent of the angiotensin-converting enzyme 2 receptor. First, via macropinocytosis, intact EVs produced from Vero E6 (monkey) cells were able to enter cells from a variety of animals, including cats, dogs, bats, hamsters, and minks, and vice versa. Second, when given to zebrafish with cutaneous wounds, the EVs showed favorable stability in aqueous environments and entered the fish. Moreover, infection of wild-type (WT) mice with heterogeneous EVs carrying SARS-CoV-2 particles led to a strong cytokine response and a notable amount of lung damage. Conversely, free viral particles did not infect WT mice. These results highlight the variety of processes behind viral transmission and cross-species evolution by indicating that EVs may be possible vehicles for SARS-CoV-2 spillover and raising risk concerns over EVs' potential for viral gene transfer.


Sujet(s)
COVID-19 , Vésicules extracellulaires , SARS-CoV-2 , Animaux , Vésicules extracellulaires/virologie , Vésicules extracellulaires/métabolisme , SARS-CoV-2/physiologie , SARS-CoV-2/pathogénicité , SARS-CoV-2/génétique , COVID-19/transmission , COVID-19/virologie , Souris , Chlorocebus aethiops , Cellules Vero , Humains , Cricetinae , Protéines d'enveloppe des coronavirus/métabolisme , Protéines d'enveloppe des coronavirus/génétique , Chiens , Danio zébré/virologie , Chats , Chiroptera/virologie , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique
11.
Bull Exp Biol Med ; 177(1): 104-108, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38960957

RÉSUMÉ

Association was assessed between the data harvested by a long-baseline laser interference deformograph and the dynamics of body temperature (BT) in hamsters deprived of natural daily light-darkness changes. The power spectral data revealed the positive correlation between simultaneous time series of hamster BT and the Earth's crust deformation (ECD). The superposed epoch analysis established an association between abrupt upstrokes of hamster BT and ECD increments. Thus, the direct relationships between BT dynamics (reflecting predominance of sympathetic part of autonomic nervous system) and ECD (according to long-baseline laser interference deformography) were established. The study observed synchronization of the free-running circadian rhythm of hamster BT with the tidal stress in Earth's lithosphere. Further studies are needed to find the physical factor underlying the revealed relationships.


Sujet(s)
Température du corps , Rythme circadien , Rythme ultradien , Animaux , Rythme ultradien/physiologie , Température du corps/physiologie , Cricetinae , Rythme circadien/physiologie , Mâle , , Mesocricetus
12.
Nat Commun ; 15(1): 5589, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38961063

RÉSUMÉ

As the new SARS-CoV-2 Omicron variants and subvariants emerge, there is an urgency to develop intranasal, broadly protective vaccines. Here, we developed highly efficacious, intranasal trivalent SARS-CoV-2 vaccine candidates (TVC) based on three components of the MMR vaccine: measles virus (MeV), mumps virus (MuV) Jeryl Lynn (JL1) strain, and MuV JL2 strain. Specifically, MeV, MuV-JL1, and MuV-JL2 vaccine strains, each expressing prefusion spike (preS-6P) from a different variant of concern (VoC), were combined to generate TVCs. Intranasal immunization of IFNAR1-/- mice and female hamsters with TVCs generated high levels of S-specific serum IgG antibodies, broad neutralizing antibodies, and mucosal IgA antibodies as well as tissue-resident memory T cells in the lungs. The immunized female hamsters were protected from challenge with SARS-CoV-2 original WA1, B.1.617.2, and B.1.1.529 strains. The preexisting MeV and MuV immunity does not significantly interfere with the efficacy of TVC. Thus, the trivalent platform is a promising next-generation SARS-CoV-2 vaccine candidate.


Sujet(s)
Administration par voie nasale , Anticorps neutralisants , Anticorps antiviraux , Vaccins contre la COVID-19 , COVID-19 , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Animaux , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , Femelle , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , COVID-19/prévention et contrôle , COVID-19/immunologie , COVID-19/virologie , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Souris , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/administration et posologie , Cricetinae , Humains , Vaccin contre la rougeole, les oreillons et la rubéole/immunologie , Vaccin contre la rougeole, les oreillons et la rubéole/administration et posologie , Virus de la rougeole/immunologie , Virus de la rougeole/génétique , Immunoglobuline G/sang , Immunoglobuline G/immunologie , Virus des oreillons/immunologie , Virus des oreillons/génétique , Souris knockout , Mesocricetus , Immunoglobuline A/immunologie , Immunoglobuline A/sang
13.
Appl Microbiol Biotechnol ; 108(1): 424, 2024 Jul 22.
Article de Anglais | MEDLINE | ID: mdl-39037584

RÉSUMÉ

Leptospirosis, a neglected zoonotic disease, is caused by pathogenic spirochetes belonging to the genus Leptospira and has one of the highest morbidity and mortality rates worldwide. Vaccination stands out as one of the most effective preventive measures for susceptible populations. Within the outer membrane of Leptospira spp., we find the LIC12287, LIC11711, and LIC13259 lipoproteins. These are of interest due to their surface location and potential immunogenicity. Thorough examination revealed the conservation of these proteins among pathogenic Leptospira spp.; we mapped the distribution of T- and B-cell epitopes along their sequences and assessed the 3D structures of each protein. This information aided in selecting immunodominant regions for the development of a chimeric protein. Through gene synthesis, we successfully constructed a chimeric protein, which was subsequently expressed, purified, and characterized. Hamsters were immunized with the chimeric lipoprotein, formulated with adjuvants aluminum hydroxide, EMULSIGEN®-D, Sigma Adjuvant System®, and Montanide™ ISA206VG. Another group was vaccinated with an inactivated Escherichia coli bacterin expressing the chimeric protein. Following vaccination, hamsters were challenged with a virulent L. interrogans strain. Our evaluation of the humoral immune response revealed the production of IgG antibodies, detectable 28 days after the second dose, in contrast to pre-immune samples and control groups. This demonstrates the potential of the chimeric protein to elicit a robust humoral immune response; however, no protection against challenge was achieved. While this study provides valuable insights into the subject, further research is warranted to identify protective antigens that could be utilized in the development of a leptospirosis vaccine. KEY POINTS: • Several T- and B-cell epitopes were identified in all the three proteins. • Four different adjuvants were used in vaccine formulations. • Immunization stimulated significant levels of IgG2/3 in vaccinated animals.


Sujet(s)
Anticorps antibactériens , Vaccins antibactériens , Leptospirose , Lipoprotéines , Animaux , Leptospirose/prévention et contrôle , Leptospirose/immunologie , Lipoprotéines/immunologie , Lipoprotéines/génétique , Vaccins antibactériens/immunologie , Vaccins antibactériens/génétique , Anticorps antibactériens/sang , Anticorps antibactériens/immunologie , Cricetinae , Déterminants antigéniques des lymphocytes B/immunologie , Déterminants antigéniques des lymphocytes B/génétique , Protéines de fusion recombinantes/immunologie , Protéines de fusion recombinantes/génétique , Adjuvants immunologiques/administration et posologie , Immunoglobuline G/sang , Déterminants antigéniques des lymphocytes T/immunologie , Déterminants antigéniques des lymphocytes T/génétique , Leptospira interrogans/immunologie , Leptospira interrogans/génétique , Protéines de la membrane externe bactérienne/immunologie , Protéines de la membrane externe bactérienne/génétique , Vaccination , Immunité humorale , Leptospira/immunologie , Leptospira/génétique , Immunogénicité des vaccins
14.
Sci Transl Med ; 16(753): eado2817, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38924429

RÉSUMÉ

The evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in variants that can escape neutralization by therapeutic antibodies. Here, we describe AZD3152, a SARS-CoV-2-neutralizing monoclonal antibody designed to provide improved potency and coverage against emerging variants. AZD3152 binds to the back left shoulder of the SARS-CoV-2 spike protein receptor binding domain and prevents interaction with the human angiotensin-converting enzyme 2 receptor. AZD3152 potently neutralized a broad panel of pseudovirus variants, including the currently dominant Omicron variant JN.1 but has reduced potency against XBB subvariants containing F456L. In vitro studies confirmed F456L resistance and additionally identified T415I and K458E as escape mutations. In a Syrian hamster challenge model, prophylactic administration of AZD3152 protected hamsters from weight loss and inflammation-related lung pathologies and reduced lung viral load. In the phase 1 sentinel safety cohort of the ongoing SUPERNOVA study (ClinicalTrials.gov: NCT05648110), a single 600-mg intramuscular injection of AZD5156 (containing 300 mg each of AZD3152 and cilgavimab) was well tolerated in adults through day 91. Observed serum concentrations of AZD3152 through day 91 were similar to those observed with cilgavimab and consistent with predictions for AZD7442, a SARS-CoV-2-neutralizing antibody combination of cilgavimab and tixagevimab, in a population pharmacokinetic model. On the basis of its pharmacokinetic characteristics, AZD3152 is predicted to provide durable protection against symptomatic coronavirus disease 2019 caused by susceptible SARS-CoV-2 variants, such as JN.1, in humans.


Sujet(s)
Anticorps neutralisants , COVID-19 , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Animaux , SARS-CoV-2/effets des médicaments et des substances chimiques , Humains , COVID-19/virologie , Anticorps neutralisants/immunologie , Glycoprotéine de spicule des coronavirus/métabolisme , Cricetinae , Traitements médicamenteux de la COVID-19 , Anticorps monoclonaux humanisés/pharmacologie , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/pharmacocinétique , Mesocricetus , Femelle , Mâle , Adulte , Anticorps antiviraux/immunologie , Mutation/génétique , Anticorps monoclonaux , Angiotensin-converting enzyme 2/métabolisme , Charge virale/effets des médicaments et des substances chimiques
15.
Methods Mol Biol ; 2810: 1-10, 2024.
Article de Anglais | MEDLINE | ID: mdl-38926269

RÉSUMÉ

We describe a method for polyethyleneimine (PEI)-mediated transient transfection of suspension-adapted Chinese hamster ovary (CHO-DG44) cells for protein expression applicable at scales from 2 mL to 2 L. The method involves transfection at a high cell density (5 × 106 cells/mL) by direct addition of plasmid DNA (pDNA) and PEI to the culture and subsequent incubation at 31 °C with agitation by orbital shaking. This method requires 0.3 mg/L of coding pDNA, 2.7 mg/L of nonspecific (filler) DNA, and 15 mg/L of PEI. The production phase is performed at 31 °C in the presence of 0.25% N,N-dimethylacetamide (DMA). If desired, the method can be modified to avoid use of DMA by increasing the amount of coding DNA. We also provide information on culture vessel options, recommended working volumes, and recommended shaking speeds for transfections at scales from 2 mL to 2 L.


Sujet(s)
Cricetulus , Plasmides , Polyéthylèneimine , Transfection , Animaux , Cellules CHO , Polyéthylèneimine/composition chimique , Transfection/méthodes , Plasmides/génétique , Expression des gènes , Cricetinae , ADN/génétique
16.
Methods Mol Biol ; 2810: 99-121, 2024.
Article de Anglais | MEDLINE | ID: mdl-38926275

RÉSUMÉ

The continuous improvement of expression platforms is necessary to respond to the increasing demand for recombinant proteins that are required to carry out structural or functional studies as well as for their characterization as biotherapeutics. While transient gene expression (TGE) in mammalian cells constitutes a rapid and well-established approach, non-clonal stably transfected cells, or "pools," represent another option, which is especially attractive when recurring productions of the same protein are required. From a culture volume of just a few liters, stable pools can provide hundreds of milligrams to gram quantities of high-quality secreted recombinant proteins.In this chapter, we describe a highly efficient and cost-effective procedure for the generation of Chinese Hamster Ovary cell stable pools expressing secreted recombinant proteins using commercially available serum-free media and polyethylenimine (PEI) as the transfection reagent. As a specific example of how this protocol can be applied, the production and downstream purification of recombinant His-tagged trimeric SARS-CoV-2 spike protein ectodomain (SmT1) are described.


Sujet(s)
Cricetulus , Polyéthylèneimine , Protéines recombinantes , Glycoprotéine de spicule des coronavirus , Transfection , Cellules CHO , Animaux , Protéines recombinantes/génétique , Protéines recombinantes/biosynthèse , Protéines recombinantes/isolement et purification , Protéines recombinantes/métabolisme , Transfection/méthodes , Polyéthylèneimine/composition chimique , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/métabolisme , Glycoprotéine de spicule des coronavirus/biosynthèse , Glycoprotéine de spicule des coronavirus/isolement et purification , SARS-CoV-2/génétique , SARS-CoV-2/métabolisme , Cricetinae , Milieux de culture sans sérum
17.
Methods Mol Biol ; 2810: 197-210, 2024.
Article de Anglais | MEDLINE | ID: mdl-38926281

RÉSUMÉ

Single-cell isolation is a key step in the manufacturing of therapeutic proteins, which relies on the development of monoclonal cell lines. It increases production safety and consistency. It also ensures higher manufacturing performances thanks to the selection of the rare clonally derived cell lines with optimal growth and production capacities. DISPENCELL-S3 is a small format single-cell dispenser whose technology is based on impedance spectroscopy. Here, we provide a detailed protocol for generating Chinese hamster ovary (CHO) monoclonal cell lines using DISPENCELL-S3. Production and characterization of an adequate cell sample for single-cell isolation, as well as the optimization of the DISPENCELL-S3 dispensing parameters are described. Monoclonal outgrowth assessment and the use of the recorded impedance signal as evidence of clonality are also outlined.


Sujet(s)
Techniques de culture cellulaire , Cricetulus , Animaux , Cellules CHO , Techniques de culture cellulaire/méthodes , Cricetinae , Séparation cellulaire/méthodes , Anticorps monoclonaux , Spectroscopie diélectrique
18.
Methods Mol Biol ; 2810: 235-247, 2024.
Article de Anglais | MEDLINE | ID: mdl-38926283

RÉSUMÉ

Most pharmaceutical biotechnology companies use stirred-tank bioreactors (STR) for recombinant protein manufacturing. These bioreactors are used at a variety of different scales ranging from bench to production scales, with working volumes from 10 mL to 25,000 L. Bench-scale STRs are commonly used to culture mammalian cells for process development, to troubleshoot production scale bioreactors using scale-down models (SDM), or to conduct fundamental research. In this chapter, we describe the operations of a bench-scale STR for the production of recombinant proteins with suspension-adapted Chinese hamster ovary (CHO) cells. These operations include bioreactor setup and configuration, batching media, inoculation of the seed cell culture, production phase, and harvest of cell-free fluids.


Sujet(s)
Bioréacteurs , Cricetulus , Protéines recombinantes , Cellules CHO , Animaux , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Cricetinae , Techniques de culture cellulaire/méthodes , Techniques de culture cellulaire en batch/méthodes , Techniques de culture cellulaire en batch/instrumentation
19.
Methods Mol Biol ; 2810: 273-284, 2024.
Article de Anglais | MEDLINE | ID: mdl-38926285

RÉSUMÉ

Chinese hamster ovary (CHO) cells are the most important mammalian expression systems to produce recombinant proteins. To ensure a proper expression of the desired molecule, it is important to monitor and adjust bioprocess parameters like oxygen concentration as well as osmolality. However, the observation of crucial cultivation parameters can be an elaborate procedure requiring lots of hands-on work. In addition, for emerging modeling approaches for bioprocesses, a model cell line responding with a measurable signal to an external influence would be highly valuable. This protocol describes in detail the procedure to generate responsive promoters reacting to limiting conditions as well as the generation of stable sensor cell lines communicating with the operator. Thereby, hypoxia and osmolality sensing response elements established in CHO cells will be utilized to trigger the expression of a minimal CMV promoter. To assess the activity of the responsive promoter in close to real time, unstable variants of GFP and BFP will be expressed, which can be analyzed via flow cytometry. Finally, an automated sampling system coupled to a fluorescence microscope enables a continuous observation of CHO cells and reports emerging limiting conditions by detecting increasing amounts of a specific fluorescent protein.


Sujet(s)
Cricetulus , Régions promotrices (génétique) , Cellules CHO , Animaux , Cricetinae , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/métabolisme , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Cytométrie en flux/méthodes , Gènes rapporteurs
20.
Methods Mol Biol ; 2810: 249-271, 2024.
Article de Anglais | MEDLINE | ID: mdl-38926284

RÉSUMÉ

Genetic engineering plays an essential role in the development of cell lines for biopharmaceutical manufacturing. Advanced gene editing tools can improve both the productivity of recombinant cell lines as well as the quality of therapeutic antibodies. Antibody glycosylation is a critical quality attribute for therapeutic biologics because the glycan patterns on the antibody fragment crystallizable (Fc) region can alter its clinical efficacy and safety as a therapeutic drug. As an example, recombinant antibodies derived from Chinese hamster ovary (CHO) cells are generally highly fucosylated; the absence of α1,6-fucose significantly enhances antibody-dependent cell-mediated cytotoxicity (ADCC) against cancer cells. This chapter describes a protocol applying clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (Cas9) approach with different formats to disrupt the α-1,6-fucosyltransferase (FUT8) gene and subsequently inhibit α-1,6 fucosylation on antibodies expressed in CHO cells.


Sujet(s)
Systèmes CRISPR-Cas , Cricetulus , Fucose , Fucosyltransferases , Édition de gène , Cellules CHO , Animaux , Édition de gène/méthodes , Fucosyltransferases/génétique , Fucosyltransferases/métabolisme , Glycosylation , Fucose/métabolisme , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Cricetinae , Humains
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE