Academic literature on the topic 'Mucolas vaccination'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the lists of relevant articles, books, theses, conference reports, and other scholarly sources on the topic 'Mucolas vaccination.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Journal articles on the topic "Mucolas vaccination"

1

Gary, Ebony N., and Michele A. Kutzler. "Defensive Driving: Directing HIV-1 Vaccine-Induced Humoral Immunity to the Mucosa with Chemokine Adjuvants." Journal of Immunology Research 2018 (December 13, 2018): 1–14. http://dx.doi.org/10.1155/2018/3734207.

Full text
Abstract:
A myriad of pathogens gain access to the host via the mucosal route; thus, vaccinations that protect against mucosal pathogens are critical. Pathogens such as HIV, HSV, and influenza enter the host at mucosal sites such as the intestinal, urogenital, and respiratory tracts. All currently licensed vaccines mediate protection by inducing the production of antibodies which can limit pathogen replication at the site of infection. Unfortunately, parenteral vaccination rarely induces the production of an antigen-specific antibody at mucosal surfaces and thus relies on transudation of systemically generated antibody to mucosal surfaces to mediate protection. Mucosa-associated lymphoid tissues (MALTs) consist of a complex network of immune organs and tissues that orchestrate the interaction between the host, commensal microbes, and pathogens at these surfaces. This complexity necessitates strict control of the entry and exit of lymphocytes in the MALT. This control is mediated by chemoattractant chemokines or cytokines which recruit immune cells expressing the cognate receptors and adhesion molecules. Exploiting mucosal chemokine trafficking pathways to mobilize specific subsets of lymphocytes to mucosal tissues in the context of vaccination has improved immunogenicity and efficacy in preclinical models. This review describes the novel use of MALT chemokines as vaccine adjuvants. Specific attention will be placed upon the use of such adjuvants to enhance HIV-specific mucosal humoral immunity in the context of prophylactic vaccination.
APA, Harvard, Vancouver, ISO, and other styles
2

Eriksson, Kristina, Marianne Quiding-Järbrink, Jacek Osek, Åke Möller, Stellan Björk, Jan Holmgren, and Cecil Czerkinsky. "Specific-Antibody-Secreting Cells in the Rectums and Genital Tracts of Nonhuman Primates following Vaccination." Infection and Immunity 66, no. 12 (December 1, 1998): 5889–96. http://dx.doi.org/10.1128/iai.66.12.5889-5896.1998.

Full text
Abstract:
ABSTRACT To determine optimal strategies to induce specific-antibody-secreting cells (specific ASC) in the rectal and vaginal mucosae, we immunized monkeys with a prototype mucosal immunogen, cholera toxin (CT), given locally or via gastric or parenteral administration. Repeated rectal or vaginal CT immunizations induced strong mucosal and systemic ASC responses. The mucosal responses were, however, confined to the immunization sites and comprised high levels of both specific antitoxin immunoglobulin A (IgA) and IgG. Large numbers of specific IgA and IgG ASC were detected in cell suspensions from dissociated genital and rectal tissues, demonstrating local accumulation of effector B cells at these sites. Intragastric immunization with CT did not per se give rise to cervicovaginal or rectal ASC responses but did prime for a rectal IgA ASC response to local booster immunization. Both rectal and vaginal immunizations also induced circulating blood IgG ASC and IgA ASC. In conclusion, these results show that local administration of antigen to the rectal or vaginal mucosa results in higher ASC responses than systemic or distant mucosal delivery. Furthermore, both the vaginal and the rectal mucosae can serve as inductive sites for systemic ASC responses. These observations should be relevant to the development of vaccines against sexually transmitted diseases such as that caused by human immunodeficiency virus.
APA, Harvard, Vancouver, ISO, and other styles
3

Tang, Jie, Larry Cai, Chuanfei Xu, Si Sun, Yuheng Liu, Joseph Rosenecker, and Shan Guan. "Nanotechnologies in Delivery of DNA and mRNA Vaccines to the Nasal and Pulmonary Mucosa." Nanomaterials 12, no. 2 (January 11, 2022): 226. http://dx.doi.org/10.3390/nano12020226.

Full text
Abstract:
Recent advancements in the field of in vitro transcribed mRNA (IVT-mRNA) vaccination have attracted considerable attention to such vaccination as a cutting-edge technique against infectious diseases including COVID-19 caused by SARS-CoV-2. While numerous pathogens infect the host through the respiratory mucosa, conventional parenterally administered vaccines are unable to induce protective immunity at mucosal surfaces. Mucosal immunization enables the induction of both mucosal and systemic immunity, efficiently removing pathogens from the mucosa before an infection occurs. Although respiratory mucosal vaccination is highly appealing, successful nasal or pulmonary delivery of nucleic acid-based vaccines is challenging because of several physical and biological barriers at the airway mucosal site, such as a variety of protective enzymes and mucociliary clearance, which remove exogenously inhaled substances. Hence, advanced nanotechnologies enabling delivery of DNA and IVT-mRNA to the nasal and pulmonary mucosa are urgently needed. Ideal nanocarriers for nucleic acid vaccines should be able to efficiently load and protect genetic payloads, overcome physical and biological barriers at the airway mucosal site, facilitate transfection in targeted epithelial or antigen-presenting cells, and incorporate adjuvants. In this review, we discuss recent developments in nucleic acid delivery systems that target airway mucosa for vaccination purposes.
APA, Harvard, Vancouver, ISO, and other styles
4

Lindholm, Catharina, Andrew Naylor, Eva-Liz Johansson, and Marianne Quiding-Järbrink. "Mucosal Vaccination Increases Endothelial Expression of Mucosal Addressin Cell Adhesion Molecule 1 in the Human Gastrointestinal Tract." Infection and Immunity 72, no. 2 (February 2004): 1004–9. http://dx.doi.org/10.1128/iai.72.2.1004-1009.2004.

Full text
Abstract:
ABSTRACT Homing of leukocytes to various tissues is dependent on the interaction between homing receptors on leukocytes and their ligands, addressins, on endothelial cells. Mucosal immunization results in homing of antigen-specific lymphocytes back to the mucosa where they first encountered the antigen. However, it is unknown whether this homing of antigen-specific cells is mediated by an altered endothelial addressin expression after vaccination. Using different immunization routes with an oral cholera vaccine, we show that the endothelial expression of mucosal addressin cell adhesion molecule 1 (MAdCAM-1) is increased in the gastric and upper small intestinal mucosae after immunization through various local routes in the upper gastrointestinal tract. In contrast, rectal immunization did not influence the levels of MAdCAM-1 in the gastric or duodenal mucosa. Furthermore, we show that MAdCAM-1 can be induced on human endothelial cells by tumor necrosis factor alpha (TNF-α) and gamma interferon. The vaccine component cholera toxin B subunit (CTB) increased MAdCAM-1 expression on endothelial cells in cultured human gastric explants, an effect that seemed to be mediated by TNF-α. In conclusion, MAdCAM-1 expression is increased in the upper gastrointestinal tract after local immunizations with a vaccine containing CTB. This strongly suggests the involvement of MAdCAM-1 in the preferential homing of mucosal lymphocytes to their original site of activation.
APA, Harvard, Vancouver, ISO, and other styles
5

González Aznar, Elizabeth, Belkis Romeu, Miriam Lastre, Caridad Zayas, Maribel Cuello, Osmir Cabrera, Yolanda Valdez, Mildrey Fariñas, and Oliver Pérez. "Mucosal and systemic immune responses induced by a single time vaccination strategy in mice." Canadian Journal of Microbiology 61, no. 8 (August 2015): 531–38. http://dx.doi.org/10.1139/cjm-2015-0063.

Full text
Abstract:
Vaccination is considered by the World Health Organization as the most cost-effective strategy for controlling infectious diseases. In spite of great successes with vaccines, many infectious diseases are still leading killers, because of the inadequate coverage of many vaccines. Several factors have been responsible: number of doses, high vaccine reactogenicity, vaccine costs, vaccination policy, among others. Contradictorily, few vaccines are of single dose and even less of mucosal administration. However, more common infections occur via mucosa, where secretory immunoglobulin A plays an essential role. As an alternative, we proposed a novel protocol of vaccination called Single Time Vaccination Strategy (SinTimVaS) by immunizing 2 priming doses at the same time: one by mucosal route and the other by parenteral route. Here, the mucosal and systemic responses induced by Finlay adjuvants (AF Proteoliposome 1 and AF Cochleate 1) implementing SinTimVaS in BALB/c mice were evaluated. One intranasal dose of AF Cochleate 1 and an intramuscular dose of AF Proteoliposome 1 adsorbed onto aluminum hydroxide, with bovine serum albumin or tetanus toxoid as model antigens, administrated at the same time, induced potent specific mucosal and systemic immune responses. Also, we demonstrated that SinTimVaS using other mucosal routes like oral and sublingual, in combination with the subcutaneous route elicits immune responses. SinTimVaS, as a new immunization strategy, could increase vaccination coverage and reduce time–cost vaccines campaigns, adding the benefits of immune response in mucosa.
APA, Harvard, Vancouver, ISO, and other styles
6

Acosta-Ramirez, Elizabeth, Zhou Xing, Reto Schwendener, and Jun Wang. "Alveolar macrophages control the quality of CD4+ T lymphocytes in the respiratory tract upon recombinant adenoviral mucosal vaccination (39.34)." Journal of Immunology 182, no. 1_Supplement (April 1, 2009): 39.34. http://dx.doi.org/10.4049/jimmunol.182.supp.39.34.

Full text
Abstract:
Abstract Infectious diseases are acquired through mucosa surfaces making mucosal vaccination important for their control. Previously, using a recombinant adenoviral-based (Ad) vaccine we have demonstrated that a single mucosal but not parenteral immunization confers protection against intracellular bacterial challenge in the lung. Here, we examined the mechanisms regulating induction of OVA-specific CD4 T-cell responses at mucosal and systemic compartments in Balb/C mice after intranasal (i.n.) or intramuscular (i.m.) vaccination with an Ad expressing OVA. We found that i.n. vaccination induced local OVA-specific T cell activation whereas i.m. vaccination induced both local and systemic T-cell activation despite of the dosage used for vaccination. Remarkably, i.n. immunization induced a characteristic proliferative kinetic different from the one induced by i.m. vaccination. Furthermore, i.n. vaccination induced a highly polarized Th1 response while i.m. immunization induced a mixed Th1/Th2 response. Of importance, in vivo depletion of alveolar macrophages led to similar T-cell proliferation kinetics and comparable cytokine production in mice receiving i.n. and i.m. vaccination. Our data suggests that alveolar macrophages provide a special microenvironment at the mucosal site, which control the quantity and quality of CD4 T memory cells and protective immunity against mucosal infectious challenges.
APA, Harvard, Vancouver, ISO, and other styles
7

Hellfritzsch and Scherließ. "Mucosal Vaccination via the Respiratory Tract." Pharmaceutics 11, no. 8 (August 1, 2019): 375. http://dx.doi.org/10.3390/pharmaceutics11080375.

Full text
Abstract:
Vaccine delivery via mucosal surfaces is an interesting alternative to parenteral vaccine administration, as it avoids the use of a needle and syringe. Mucosal vaccine administration also targets the mucosal immune system, which is the largest lymphoid tissue in the human body. The mucosal immune response involves systemic, antigen-specific humoral and cellular immune response in addition to a local response which is characterised by a predominantly cytotoxic T cell response in combination with secreted IgA. This antibody facilitates pathogen recognition and deletion prior to entrance into the body. Hence, administration via the respiratory mucosa can be favoured for all pathogens which use the respiratory tract as entry to the body, such as influenza and for all diseases directly affecting the respiratory tract such as pneumonia. Additionally, the different mucosal tissues of the human body are interconnected via the so-called “common mucosal immune system”, which allows induction of an antigen-specific immune response in distant mucosal sites. Finally, mucosal administration is also interesting in the area of therapeutic vaccination, in which a predominant cellular immune response is required, as this can efficiently be induced by this route of delivery. The review gives an introduction to respiratory vaccination, formulation approaches and application strategies.
APA, Harvard, Vancouver, ISO, and other styles
8

Lu, Bin, Wenbo Yu, Xiaoxing Huang, Haibo Wang, Li Liu, and Zhiwei Chen. "Mucosal Immunization Induces a Higher Level of Lasting Neutralizing Antibody Response in Mice by a Replication-Competent Smallpox Vaccine: Vaccinia Tiantan Strain." Journal of Biomedicine and Biotechnology 2011 (2011): 1–9. http://dx.doi.org/10.1155/2011/970424.

Full text
Abstract:
The possible bioterrorism threat using the variola virus, the causative agent of smallpox, has promoted us to further investigate the immunogenicity profiles of existing vaccines. Here, we study for the first time the immunogenicity profile of a replication-competent smallpox vaccine (vaccinia Tiantan, VTT strain) for inducing neutralizing antibodies (Nabs) through mucosal vaccination, which is noninvasive and has a critical implication for massive vaccination programs. Four different routes of vaccination were tested in parallel including intramuscular (i.m.), intranasal (i.n.), oral (i.o.), and subcutaneous (s.c.) inoculations in mice. We found that one time vaccination with an optimal dose of VTT was able to induce anti-VTT Nabs via each of the four routes. Higher levels of antiviral Nabs, however, were induced via the i.n. and i.o. inoculations when compared with the i.m. and s.c. routes. Moreover, the i.n. and i.o. vaccinations also induced higher sustained levels of Nabs overtime, which conferred better protections against homologous or alternating mucosal routes of viral challenges six months post vaccination. The VTT-induced immunity via all four routes, however, was partially effective against the intramuscular viral challenge. Our data have implications for understanding the potential application of mucosal smallpox vaccination and for developing VTT-based vaccines to overcome preexisting antivaccinia immunity.
APA, Harvard, Vancouver, ISO, and other styles
9

Reljic, Rajko, Laura Sibley, Jen-Min Huang, Ilaria Pepponi, Andreas Hoppe, Huynh A. Hong, and Simon M. Cutting. "Mucosal Vaccination against Tuberculosis Using Inert Bioparticles." Infection and Immunity 81, no. 11 (August 19, 2013): 4071–80. http://dx.doi.org/10.1128/iai.00786-13.

Full text
Abstract:
ABSTRACTNeedle-free, mucosal immunization is a highly desirable strategy for vaccination against many pathogens, especially those entering through the respiratory mucosa, such asMycobacterium tuberculosis. Unfortunately, mucosal vaccination against tuberculosis (TB) is impeded by a lack of suitable adjuvants and/or delivery platforms that could induce a protective immune response in humans. Here, we report on a novel biotechnological approach for mucosal vaccination against TB that overcomes some of the current limitations. This is achieved by coating protective TB antigens onto the surface of inert bacterial spores, which are then delivered to the respiratory tract. Our data showed that mice immunized nasally with coated spores developed humoral and cellular immune responses and multifunctional T cells and, most importantly, presented significantly reduced bacterial loads in their lungs and spleens following pathogenic challenge. We conclude that this new vaccine delivery platform merits further development as a mucosal vaccine for TB and possibly also other respiratory pathogens.
APA, Harvard, Vancouver, ISO, and other styles
10

Feng, Fengling, Ziyu Wen, Jiaoshan Chen, Yue Yuan, Congcong Wang, and Caijun Sun. "Strategies to Develop a Mucosa-Targeting Vaccine against Emerging Infectious Diseases." Viruses 14, no. 3 (March 3, 2022): 520. http://dx.doi.org/10.3390/v14030520.

Full text
Abstract:
Numerous pathogenic microbes, including viruses, bacteria, and fungi, usually infect the host through the mucosal surfaces of the respiratory tract, gastrointestinal tract, and reproductive tract. The mucosa is well known to provide the first line of host defense against pathogen entry by physical, chemical, biological, and immunological barriers, and therefore, mucosa-targeting vaccination is emerging as a promising strategy for conferring superior protection. However, there are still many challenges to be solved to develop an effective mucosal vaccine, such as poor adhesion to the mucosal surface, insufficient uptake to break through the mucus, and the difficulty in avoiding strong degradation through the gastrointestinal tract. Recently, increasing efforts to overcome these issues have been made, and we herein summarize the latest findings on these strategies to develop mucosa-targeting vaccines, including a novel needle-free mucosa-targeting route, the development of mucosa-targeting vectors, the administration of mucosal adjuvants, encapsulating vaccines into nanoparticle formulations, and antigen design to conjugate with mucosa-targeting ligands. Our work will highlight the importance of further developing mucosal vaccine technology to combat the frequent outbreaks of infectious diseases.
APA, Harvard, Vancouver, ISO, and other styles

Dissertations / Theses on the topic "Mucolas vaccination"

1

Heritage, Philippa Louise. "Mucosal vaccination with a novel microparticle delivery system." Thesis, National Library of Canada = Bibliothèque nationale du Canada, 1998. http://www.collectionscanada.ca/obj/s4/f2/dsk1/tape11/PQDD_0003/NQ42849.pdf.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Vento, Kevin Leon. "Assessment of protective immunity following mucosal vaccination with Pseudomonas aeruginosa." Thesis, St George's, University of London, 2003. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.408031.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Moors, Adam. "Antibody mediated mucosal defences in the female genital tract." Thesis, University of Southampton, 2000. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.368058.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Rahman, Muhammad Jubayer. "Mucosal immunity against mycobacterial infection." Doctoral thesis, Stockholms universitet, Wenner-Grens institut, 2010. http://urn.kb.se/resolve?urn=urn:nbn:se:su:diva-39170.

Full text
Abstract:
This thesis aimed to the identification of immune biomarkers of mycobacterial infection for better diagnosis of tuberculosis (TB) and also focused on new vaccination strategies with a particular emphasis on the immune responses in the respiratory tract using murine models. Since the lung is the natural habitat for the M. tuberculosis, we reasoned that immune responses detected locally in the lungs would be good correlates of infection (Paper I). Likewise, immune responses induced in the respiratory tract following immunization would be more effective against mycobacterial infection. We showed that cytokines (IL-12, TNF, and IFN-γ) and cytokine receptors (sTNFR1 and sTNFR2) together with specific antibodies in the respiratory tract correlated better with the bacterial burden in the organs. In Paper II, we investigated the role of the BCG vaccination as a priming vaccine in a heterologous prime-boost immunization protocol. The results showed that the neonatal BCG vaccination primed the immune system for a relevant antigen and showed a generalized adjuvant effect. Using this immunization protocol, protective immune responses in the lungs were generated independently of the route used for the booster immunization. In Paper III, We showed that exposure to mycobacterial antigens during the gestational period led to antigen transportation from the mother to the fetus and this resulted in an early priming of the fetal immune system. Immunization with the same antigen during the postnatal life increased antigen-specific recall IFN-γ responses and protection against infection. We examined the role of innate immunity for the induction of acquired immune responses upon immunization with mycobacterial antigens using TLR2 deficient mice (Paper IV). Our data indicated that suboptimal innate immune responses in the TLR2-/- mice might compromise the induction of acquired immune responses. Overall, the current findings suggested that a better understanding of the mucosal immunity would be useful for the improvement of diagnostic procedures and the development of efficient vaccines against TB.
At the time of the doctoral defense, the following paper was unpublished and had a status as follows: Paper 4: Manuscript
APA, Harvard, Vancouver, ISO, and other styles
5

Easeman, Richard. "Induction of mucosal immune responses in the horse." Thesis, Open University, 1997. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.389312.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Hall, L. J. "Phenotypic and functional characterisation of innate and adaptive immune responses after mucosal vaccination." Thesis, University of Cambridge, 2007. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.599868.

Full text
Abstract:
Here I examine the properties of a live Salmonella-based vaccine and a mucosal adjuvant based on a bacterial protein. Initially I examined the immunogenicity of the M. tuberculosis fusion antigen Ag85B-ESAT6 using a number of different mucosal vaccination strategies. These strategies included (i) intranasal immunisation with Ag85B-ESAT6 protein with and without Heat Labile toxin as an adjuvant (ii) oral immunisation with Salmonella enterica Typhimurium expressing Ag85B-ESAT6 from in vivo inducible or constitutive promoters (with and without intranasal boosts). Mice immunised with the various vaccine candidates were found to have significant anti-Ag85B-ESAT6 serum and mucosal antibody titres as well as strong TH1 type cytokine responses, with IFN-γ levels particularly high. Intranasal boosting served to further enhance these immune responses. Following vaccination with the constitutive Salmonella vector, mice challenged with M. tuberculosis were found to have significantly reduced CFU in the liver when compared to non-vaccinated animals. Mice primed with Salmonella and then boosted intranasally with Ag85B-ESAT6/LTK63 led to a significant increase in protection, equivalent to that observed in mice vaccinated with BCG. In a separate study, flow cytometry and confocal microscopy were used to examine the frequencies and localisation of innate immune cells, their activation status, as well as the expression of cell adhesion molecules following intranasal immunisation. I found striking differences between the cell surface phenotype of leukocytes and their pattern of distribution in the tissues examined at all time points tested after immunisation. Following on from these results one particular cell type was examined in more depth to determine its role in adaptive immune responses.
APA, Harvard, Vancouver, ISO, and other styles
7

Falkeborn, Tina. "Nasal vaccination using novel mucosal adjuvants : with main focus on influenza A virus." Doctoral thesis, Linköpings universitet, Avdelningen för mikrobiologi och molekylär medicin, 2015. http://urn.kb.se/resolve?urn=urn:nbn:se:liu:diva-117981.

Full text
Abstract:
Influenza viruses have sporadically caused pandemics during the last century, with the most severe occurring in 1918 when the “Spanish flu”, an A/H1N1 influenza virus, passed around the globe killing about 20-100 million people. Today 250 000-500 000 deaths occur annually due to influenza virus or secondary infection after influenza, e.g. pneumonia. Influenza viruses cause severe infections in susceptible age groups like children and elderly and in individuals with impaired immune response due to other medical conditions. The best way to prevent an influenza epidemic is by vaccination. Since the 1950´s we have vaccines against seasonal flu, but vaccine efficacy is not 100 % and there is a need to develop better and more effective vaccines, especially for the risk groups. Since the virus enters the host through the nasal cavity, nasal vaccination is a good approach. By stimulating a mucosal immune response already in the nasal cavity, the goal with nasal vaccination is to stop the virus before it enters the host. Nasal vaccination also reduces the risk of transmission of blood-borne diseases, and is less painful and easier to administer, compared to injectable vaccines. In order to be able to use less immunogenic antigens, like split and subunit antigens, as nasal vaccine components, an adjuvant is needed to enhance the immune response. At the moment there is no licensed mucosal adjuvant for human use. Several studies are ongoing, but it is a complicated and long way to reach the market. In this thesis nasal vaccination with influenza antigen together with the mucosal adjuvant Endocine™ and other mucosal adjuvants has been evaluated. The Endocine™ adjuvant has been shown to be safe and well tolerated in clinical trials. Depending on the pathogen of interest, different approaches are necessary. For HIV, DNA-vaccination has been evaluated together with a plasmid encoding Salmonella typhimurium flagellin C and the mucosal adjuvant N3. The results found in paper I-IV show that by adding adjuvant to the antigen enhances the protective immune response towards the antigen. Enhanced systemic, mucosal and cell-mediated immunity were observed. Hopefully in the future these adjuvants evaluated in this thesis, will be used in vaccines for humans.
APA, Harvard, Vancouver, ISO, and other styles
8

Vazquez, Thomas. "Evaluation préclinique d'un protocole vaccinal anti-VIH utilisant des pseudo-particules rétrovirales recombinantes administrées par voies muqueuses et étude des mécanismes immunologiques associés." Thesis, Paris 6, 2016. http://www.theses.fr/2016PA066321.

Full text
Abstract:
Malgré 30 ans de recherche aucun vaccin VIH n'a permis d'apporter une protection efficace et de manière stable. Au regard des échecs obtenus jusqu'à présent, de nouvelles formes vaccinales ont été développées. Parmi ces dernières les VLP présentent l'avantage notables d'être très immunogènes du fait de leur forme particulaire mimant les virus natifs, et sécuritaire puisque ne véhiculant pas de génome viral. Ce travail de thèse a pour objectif d’établir et d’évaluer une stratégie vaccinale utilisant ces VLP administrées par voies muqueuses dans le but d’initier une réponse humorale et cellulaire au niveau systémique et muqueux. Dans cette étude, nous avons montré que la voie muqueuse est indispensable pour l’induction d’une réponse locale forte. De plus, nos résultats révèlent que la forme particulaire de l’antigène est décisive dans la génération d’une immunité de qualité, générant une réponse TFH forte, une réponse cellulaire locale polyfonctionnelle ainsi qu’une réponse humorale forte et stable dans le temps, caractérisée par des anticorps de qualité.Cherchant à mieux caractériser la stratégie vaccinale établie, nous avons analysé les mécanismes de prise en charge des VLP et d’initiation de la réponse immunitaire après administration IN. Nous avons observé que de nombreuses cellules de l’immunité innée pulmonaire, notamment les macrophages alvéolaires et les neutrophiles, captaient massivement les VLP limitant alors la réponse TFH et potentiellement la réponse humorale qui en découle. Au final, ce travail de thèse aura permis de mettre en avant la voie d’immunisation muqueuse ainsi que la forme particulaire de l’antigène dans la mise en place d’un vaccin VIH
Currently no HIV vaccine elicit full and stable protection against viral acquisition. In view of the failures until now, new vaccines strategies were developed. Among these, VLP have the advantage to be highly immunogenic because of their particulate structure mimicking native pathogens and safe because of the lack of viral genome.This thesis work aims to develop and evaluate a VLP-based vaccine strategy by mucosal administration in order to initiate systemic and local humoral and cellular responses. In this study, we showed that the mucosal administration is mandatory to generate a strong local immunity. Moreover, our results show that particular form of the antigen is crucial in the generation of the quality of the immune response, generating strong TFH response, polyfunctional T-cell responses in the mucosa and a strong and stable humoral response characterized by high-quality antibodies.We also investigated mechanisms involved in the generation of immune responses following IN VLP injections. We determined which cells take in charge VLP and their role in the followed immune responses. Our preliminary results show many innate immune cells in the lungs, such as alveolar macrophages and neutrophils, have an important role in the particles uptake, limiting TFH response and potentially the followed humoral response.Finally, this thesis work will show the determining role of the mucosal route of immunization and the particulate form antigen for the development of an HIV vaccine
APA, Harvard, Vancouver, ISO, and other styles
9

Biswal, Jitendra Kumar. "Evaluation of mucosal immunity in FMDV vaccinated and infected cattle." Thesis, Royal Veterinary College (University of London), 2012. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.572448.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

McGuire, Carolann. "Mucosal vaccination using a crude antigen and a synthetic peptide in the Trichinella spiralis model." Thesis, University of Nottingham, 1998. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.285567.

Full text
APA, Harvard, Vancouver, ISO, and other styles

Books on the topic "Mucolas vaccination"

1

Kozlowski, Pamela A. Mucosal Vaccines: Modern Concepts, Strategies, and Challenges. Berlin, Heidelberg: Springer Berlin Heidelberg, 2012.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
2

Frey, Andreas, Pam Kozlowski, and Nicholas J. Mantis, eds. Mucosal Vaccination: Strategies to Induce and Evaluate Mucosal Immunity. Frontiers Media SA, 2022. http://dx.doi.org/10.3389/978-2-88976-216-3.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Mucosal Vaccines Current Topics in Microbiology and Immmunology. Springer, 2012.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
4

Kozlowski, Pamela A. Mucosal Vaccines: Modern Concepts, Strategies, and Challenges. Springer, 2014.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
5

E, Ciardi J., McGhee Jerry R, Keith Jerry M, and National Institute of Dental Research (U.S.), eds. Genetically engineered vaccines. New York: Plenum Press, 1992.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
6

(Editor), Joseph E. Ciardi, Jerry R. McGhee (Editor), and Jerry M. Keith (Editor), eds. Genetically Engineered Vaccines (Advances in Experimental Medicine and Biology). Springer, 1993.

Find full text
APA, Harvard, Vancouver, ISO, and other styles

Book chapters on the topic "Mucolas vaccination"

1

Rombout, Jan H. W. M., and Viswanath Kiron. "Mucosal Vaccination of Fish." In Fish Vaccination, 56–67. Chichester, UK: John Wiley & Sons, Ltd, 2014. http://dx.doi.org/10.1002/9781118806913.ch6.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Borges, Olga, and Gerrit Borchard. "Mucosal Vaccination: Opportunities and Challenges." In Novel Immune Potentiators and Delivery Technologies for Next Generation Vaccines, 65–80. Boston, MA: Springer US, 2012. http://dx.doi.org/10.1007/978-1-4614-5380-2_3.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

García-Hernández, Ana Lilia, Néstor Rubio-Infante, and Leticia Moreno-Fierros. "Mucosal Immunology and Oral Vaccination." In Genetically Engineered Plants as a Source of Vaccines Against Wide Spread Diseases, 15–42. New York, NY: Springer New York, 2014. http://dx.doi.org/10.1007/978-1-4939-0850-9_2.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Borchard, Gerrit, Farnaz Esmaeili, and Simon Heuking. "Chitosan-Based Delivery Systems for Mucosal Vaccination." In Chitosan-Based Systems for Biopharmaceuticals, 211–24. Chichester, UK: John Wiley & Sons, Ltd, 2012. http://dx.doi.org/10.1002/9781119962977.ch12.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Watts, Peter, Alan Smith, and Michael Hinchcliffe. "ChiSys® as a Chitosan-Based Delivery Platform for Nasal Vaccination." In Mucosal Delivery of Biopharmaceuticals, 499–516. Boston, MA: Springer US, 2014. http://dx.doi.org/10.1007/978-1-4614-9524-6_23.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Kantele, A. "Antibody secreting cells after oral vaccination with live Salmonella typhi vaccines." In Advances in Mucosal Immunology, 355–58. Dordrecht: Springer Netherlands, 1990. http://dx.doi.org/10.1007/978-94-009-1848-1_98.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Campo, M. Saveria, and William F. H. Jarrett. "Vaccination Against Cutaneous and Mucosal Papillomavirus in Cattle." In Ciba Foundation Symposium 187 - Vaccines Against Virally Induced Cancers, 61–77. Chichester, UK: John Wiley & Sons, Ltd., 2007. http://dx.doi.org/10.1002/9780470514672.ch5.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Kantele, A., H. Arvilommi, A. Ratilainen, L. Rintala, J. M. Kantele, and P. H. Mäkelä. "Antibody-secreting cell responses after vaccination with parenteral killed, oral killed or oral live vaccine." In Advances in Mucosal Immunology, 353–54. Dordrecht: Springer Netherlands, 1990. http://dx.doi.org/10.1007/978-94-009-1848-1_97.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Trudel, M., F. Nadon, C. Séguin, and P. Talbot. "Efficient intranasal vaccination of mice against human respiratory syncytial virus with an experimental iscoms subunit vaccine." In Advances in Mucosal Immunology, 379–81. Dordrecht: Springer Netherlands, 1990. http://dx.doi.org/10.1007/978-94-009-1848-1_105.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Fujihashi, Kohtaro, Jerry R. McGhee, and Hiroshi Kiyono. "Mucosal Vaccination Challenges in Aging: Understanding Immunosenescence in the Aerodigestive Tract." In Handbook of Immunosenescence, 1379–405. Cham: Springer International Publishing, 2019. http://dx.doi.org/10.1007/978-3-319-99375-1_114.

Full text
APA, Harvard, Vancouver, ISO, and other styles

Conference papers on the topic "Mucolas vaccination"

1

Pereira, Igor Muzetti, and Vinícius Victor Lelis. "Developing a open-source serious game for control and education on HPV and Cervical Cancer." In Congresso Latino-Americano de Software Livre e Tecnologias Abertas. Sociedade Brasileira de Computação - SBC, 2019. http://dx.doi.org/10.5753/latinoware.2019.10344.

Full text
Abstract:
HPV - short for human papillomavirus - is a virus capable of infecting the mucous membranes and skin, being the most prevalent involved on Sexually Transmitted Diseases and the main cause of Cervical Cancer. However, reaching out to and inviting all eligible people for cervical cancer screening and vaccination against HPV is a difficult task. A possible solution is the use of social inducement and gamification through modern means of communication, which may encourage the attendance of unscreened or unvaccinated people on related programs. Therefore, this paper presents the development of a serious game for the Android platform designed for children and preteens with the purpose of foresting HPV and Cervical Cancer awareness, informationseeking, and communication, thus possibly increasing the number of vaccinated and screened people.
APA, Harvard, Vancouver, ISO, and other styles
2

Bexiga, Natalia, Celso Caricati, Marcos Capone, Adriano Alencar, and Marco Stephano. "Carboxymethyl chitosan (CMCS) nanoparticles for mucosal vaccination against rabies: evaluation of the immune response following oral immunization studies in mice." In IV International Symposium on Immunobiologicals & VII Seminário Anual Científico e Tecnológico. Instituto de Tecnologia em Imunobiológicos, 2019. http://dx.doi.org/10.35259/isi.sact.2019_32527.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Oliveira, Raddib Eduardo Noleto da Nobrega, Rafael Pereira Guimarães, Maria Eduarda Angelo de Mendonça Filleti, and Thábata Emanuelle Martins Nunes. "Optical neurorretinitis by Bartonella Henselae - case report." In XIII Congresso Paulista de Neurologia. Zeppelini Editorial e Comunicação, 2021. http://dx.doi.org/10.5327/1516-3180.535.

Full text
Abstract:
Introduction: Cat scratch disease is an infection caused by Bartonella henselae, usually transmitted to humans through cat scratch or bite. The most common clinical manifestation is lymphadenitis, but 5 to 10% of patients with cat scratch disease may have ocular involvement. Objectives and methodology: This work aims to report a case of Neuroretinitis optica by Bartonella Henselae. Data were collected through analysis of medical records with the patient’s consent. Results / Case report: Female, 22 years old, born in Joinville, presented, acutely, decreased visual acuity in RE, without pain on eye movement. She reported flu with fever in the previous month and denied recent vaccination. Visual acuity was 20/40 in RE and the fundus revealed papilla edema, hemorrhage and uveitis (+ / 4 +). In laboratory tests there were no changes. Serology for toxoplasmosis revealed a slight increase in IgM and the other serologies were negative. Sulfamethoxazole 800 mg / trimethoprim 160 mg started 12/12 hs and prednisone 80 mg / day, without improvement. Evolved with worsening and visual acuity (20/100) in OD. Retinography showed vascular narrowing, papillary blurring, decreased foveal brightness and macular edema, configuring optic neuritis D, with no changes in the LE. The neurological evaluation did not find any findings other than visual changes. The CSF study, cranial and orbit MRs were normal. At that time, the patient reported having had contact with a dead kitten. Serology was positive for Bartonella (IgM 1/100). Doxycycline 100mg started at 12 / 12h. After 15 days, a stellate macula and a slight improvement in papilla edema were observed. The patient evolved with full recovery. Conclusion: B.henselae is the main etiological agent of DAG. Kittens are the main reservoirs of B.henselae. Contact with mucous membranes or conjunctivae may be involved. Cat scratch disease (GAD) has two clinical presentations. Typical GAD is characterized by subacute regional lymphadenopathy; atypical GAD is the designation for numerous manifestations involving several organs, and occurs in 10- 15% of cases, being responsible for Parinaud’s oculoglandular syndrome.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography