1
|
Priyanka, Abusalah MAH, Chopra H, Sharma A, Mustafa SA, Choudhary OP, Sharma M, Dhawan M, Khosla R, Loshali A, Sundriyal A, Saini J. Nanovaccines: A game changing approach in the fight against infectious diseases. Biomed Pharmacother 2023; 167:115597. [PMID: 37783148 DOI: 10.1016/j.biopha.2023.115597] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023] Open
Abstract
The field of nanotechnology has revolutionised global attempts to prevent, treat, and eradicate infectious diseases in the foreseen future. Nanovaccines have proven to be a valuable pawn in this novel technology. Nanovaccines are made up of nanoparticles that are associated with or prepared with components that can stimulate the host's immune system. In addition to their delivery capabilities, the nanocarriers have been demonstrated to possess intrinsic adjuvant properties, working as immune cell stimulators. Thus, nanovaccines have the potential to promote rapid as well as long-lasting humoral and cellular immunity. The nanovaccines have several possible benefits, including site-specific antigen delivery, increased antigen bioavailability, and a diminished adverse effect profile. To avail these benefits, several nanoparticle-based vaccines are being developed, including virus-like particles, liposomes, polymeric nanoparticles, nanogels, lipid nanoparticles, emulsion vaccines, exomes, and inorganic nanoparticles. Inspired by their distinctive properties, researchers are working on the development of nanovaccines for a variety of applications, such as cancer immunotherapy and infectious diseases. Although a few challenges still need to be overcome, such as modulation of the nanoparticle pharmacokinetics to avoid rapid elimination from the bloodstream by the reticuloendothelial system, The future prospects of this technology are also assuring, with multiple options such as personalised vaccines, needle-free formulations, and combination nanovaccines with several promising candidates.
Collapse
Affiliation(s)
- Priyanka
- Department of Veterinary Microbiology, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Rampura Phul, Bathinda 151103, Punjab, India
| | - Mai Abdel Haleem Abusalah
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Zarqa University, Al-Zarqa 13132, Jordan
| | - Hitesh Chopra
- Department of Biosciences, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Abhilasha Sharma
- Department of Life Science, Gujarat University, University School of Sciences, Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Suhad Asad Mustafa
- Scientific Research Center/ Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Om Prakash Choudhary
- Department of Veterinary Anatomy, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Rampura Phul, Bathinda 151103, Punjab, India.
| | - Manish Sharma
- University Institute of Biotechnology, Department of Biotechnology, Chandigarh University, Mohali 140413, Punjab, India
| | - Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana 141004, Punjab, India; Trafford College, Altrincham, Manchester WA14 5PQ, UK.
| | - Rajiv Khosla
- Department of Biotechnology, Doaba College, Jalandhar 144004, Punjab, India
| | - Aanchal Loshali
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ankush Sundriyal
- School of Pharmaceutical Sciences and Research, Sardar Bhagwan Singh University, Balawala, Dehradun 248001, India
| | - Jyoti Saini
- Department of Veterinary Anatomy, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Rampura Phul, Bathinda 151103, Punjab, India
| |
Collapse
|
2
|
Seyfoori A, Shokrollahi Barough M, Mokarram P, Ahmadi M, Mehrbod P, Sheidary A, Madrakian T, Kiumarsi M, Walsh T, McAlinden KD, Ghosh CC, Sharma P, Zeki AA, Ghavami S, Akbari M. Emerging Advances of Nanotechnology in Drug and Vaccine Delivery against Viral Associated Respiratory Infectious Diseases (VARID). Int J Mol Sci 2021; 22:6937. [PMID: 34203268 PMCID: PMC8269337 DOI: 10.3390/ijms22136937] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/19/2021] [Accepted: 06/19/2021] [Indexed: 12/12/2022] Open
Abstract
Viral-associated respiratory infectious diseases are one of the most prominent subsets of respiratory failures, known as viral respiratory infections (VRI). VRIs are proceeded by an infection caused by viruses infecting the respiratory system. For the past 100 years, viral associated respiratory epidemics have been the most common cause of infectious disease worldwide. Due to several drawbacks of the current anti-viral treatments, such as drug resistance generation and non-targeting of viral proteins, the development of novel nanotherapeutic or nano-vaccine strategies can be considered essential. Due to their specific physical and biological properties, nanoparticles hold promising opportunities for both anti-viral treatments and vaccines against viral infections. Besides the specific physiological properties of the respiratory system, there is a significant demand for utilizing nano-designs in the production of vaccines or antiviral agents for airway-localized administration. SARS-CoV-2, as an immediate example of respiratory viruses, is an enveloped, positive-sense, single-stranded RNA virus belonging to the coronaviridae family. COVID-19 can lead to acute respiratory distress syndrome, similarly to other members of the coronaviridae. Hence, reviewing the current and past emerging nanotechnology-based medications on similar respiratory viral diseases can identify pathways towards generating novel SARS-CoV-2 nanotherapeutics and/or nano-vaccines.
Collapse
Affiliation(s)
- Amir Seyfoori
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (T.W.)
- Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran 1517964311, Iran
| | - Mahdieh Shokrollahi Barough
- Department of Immunology, Iran University of Medical Sciences, Tehran 1449614535, Iran;
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran 1517964311, Iran
| | - Pooneh Mokarram
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran;
- Autophagy Research Center, Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Mazaher Ahmadi
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (M.A.); (T.M.)
| | - Parvaneh Mehrbod
- Influenza and Respiratory Viruses Department, Pasteur Institute of IRAN, Tehran 1316943551, Iran;
| | - Alireza Sheidary
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14155-6451, Iran;
| | - Tayyebeh Madrakian
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (M.A.); (T.M.)
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14155-6451, Iran;
| | - Mohammad Kiumarsi
- Department of Human Anatomy and Cell Science, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
| | - Tavia Walsh
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (T.W.)
| | - Kielan D. McAlinden
- Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS 7248, Australia;
| | - Chandra C. Ghosh
- Roger Williams Medical Center, Immuno-Oncology Institute (Ix2), Providence, RI 02908, USA;
| | - Pawan Sharma
- Center for Translational Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Amir A. Zeki
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, U.C. Davis Lung Center, Davis School of Medicine, University of California, Davis, CA 95817, USA;
- Veterans Affairs Medical Center, Mather, CA 95817, USA
| | - Saeid Ghavami
- Autophagy Research Center, Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Department of Human Anatomy and Cell Science, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Mohsen Akbari
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (T.W.)
- Biotechnology Center, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland
- Center for Advanced Materials and Related Technologies, University of Victoria, Victoria, BC V8P 5C2, Canada
| |
Collapse
|
3
|
Xyloglucan based mucosal nanovaccine for immunological protection against brucellosis developed by supercritical fluid technology. INTERNATIONAL JOURNAL OF PHARMACEUTICS-X 2020; 2:100053. [PMID: 32776000 PMCID: PMC7397708 DOI: 10.1016/j.ijpx.2020.100053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 06/28/2020] [Accepted: 07/13/2020] [Indexed: 11/21/2022]
Abstract
Vaccines delivered via the mucosal route have logistic benefits over parenteral or intramuscular vaccines as they offer patient compliance. This study presents the first intranasal, controlled release, subunit nanovaccine comprising mucoadhesive tamarind seed polymer (xyloglucan) based nanoparticles produced using an efficient, environmentally compatible, and industrially scalable technique: rapid expansion of supercritical solution. The nanovaccine formulation aimed against brucellosis comprised xyloglucan nanoparticles loaded separately with antigenic acellular lipopolysaccharides from B. abortus (S19) and the immunoadjuvant quillaja saponin. The nanovaccine elicited prolonged humoral and cell-mediated immunity in female Balb/c mice. Nasal vaccination with the nanovaccine resulted in higher levels of mucosal IgA and IgG than with an aqueous solution of soluble lipopolysaccharides and quillaja saponin. Systemic immunity triggered by the nanovaccine was evidenced by higher IgG levels in sera post priming and boosting. The nanovaccine induced a mixed Th1/Th2 type of immunity with higher IgG2a levels and thus a polarized Th1 response. The results suggest that the nanovaccine administered by homologous nasal route can prime the immune system via the mucosal and systemic pathways and is a good candidate for vaccine delivery.
Collapse
|
4
|
Mucosal Vaccination via the Respiratory Tract. Pharmaceutics 2019; 11:pharmaceutics11080375. [PMID: 31374959 PMCID: PMC6723941 DOI: 10.3390/pharmaceutics11080375] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/12/2019] [Accepted: 07/23/2019] [Indexed: 12/11/2022] Open
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.
Collapse
|
5
|
Carrasco-Yepez MM, Campos-Rodríguez R, Reséndiz-Albor AA, Peña-Juárez C, Contis-Montes de Oca A, Arciniega-Martínez IM, Bonilla-Lemus P, Rojas-Hernandez S. Naegleria fowleri immunization modifies lymphocytes and APC of nasal mucosa. Parasite Immunol 2018; 40. [PMID: 29243267 DOI: 10.1111/pim.12508] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/01/2017] [Indexed: 11/28/2022]
Abstract
We investigated whether intranasal immunization with amoebic lysates plus cholera toxin modified the populations of T and B lymphocytes, macrophages and dendritic cells by flow cytometry from nose-associated lymphoid tissue (NALT), cervical lymph nodes (CN), nasal passages (NP) and spleen (SP). In all immunized groups, the percentage of CD4 was higher than CD8 cells. CD45 was increased in B cells from mice immunized. We observed IgA antibody-forming cell (IgA-AFC) response, mainly in NALT and NP. Macrophages from NP and CN expressed the highest levels of CD80 and CD86 in N. fowleri lysates with either CT or CT alone immunized mice, whereas dendritic cells expressed high levels of CD80 and CD86 in all compartment from immunized mice. These were lower than those expressed by macrophages. Only in SP from CT-immunized mice, these costimulatory molecules were increased. These results suggest that N. fowleri and CT antigens are taking by APCs, and therefore, protective immunity depends on interactions between APCs and T cells from NP and CN. Consequently, CD4 cells stimulate the differentiation from B lymphocytes to AFC IgA-positive; antibody that we previously found interacting with trophozoites in the nasal lumen avoiding the N. fowleri attachment to nasal epithelium.
Collapse
Affiliation(s)
- M M Carrasco-Yepez
- Proyecto CyMA, Los Reyes Iztacala, UIICSE, UNAM FES Iztacala, Tlalnepantla, Mexico
| | - R Campos-Rodríguez
- Departamento de Bioquímica, Escuela Superior de Medicina, Laboratorio de Bioquímica, Instituto Politécnico Nacional, Mexico City, Mexico
| | - A A Reséndiz-Albor
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Laboratorio de Inmunobiología Molecular y Celular, Instituto Politécnico Nacional, Mexico City, Mexico
| | - C Peña-Juárez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Laboratorio de Inmunobiología Molecular y Celular, Instituto Politécnico Nacional, Mexico City, Mexico
| | - A Contis-Montes de Oca
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Laboratorio de Inmunobiología Molecular y Celular, Instituto Politécnico Nacional, Mexico City, Mexico
| | - I M Arciniega-Martínez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Laboratorio de Inmunobiología Molecular y Celular, Instituto Politécnico Nacional, Mexico City, Mexico
| | - P Bonilla-Lemus
- Proyecto CyMA, Los Reyes Iztacala, UIICSE, UNAM FES Iztacala, Tlalnepantla, Mexico
| | - S Rojas-Hernandez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Laboratorio de Inmunobiología Molecular y Celular, Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
6
|
Bankvall M, Jontell M, Wold A, Östman S. Tissue-specific Differences in Immune Cell Subsets Located in the Naso-oropharyngeal-associated Lymphoid Tissues. Scand J Immunol 2017; 87:15-27. [PMID: 29077981 DOI: 10.1111/sji.12625] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 10/21/2017] [Indexed: 01/09/2023]
Abstract
Defining the immune cells within the naso-oropharyngeal-associated lymphoid tissues would promote the development of efficient orally and nasally delivered immunotherapies. The aim was to compare murine antigen-presenting cells (APCs) and T cell subsets in the nose-associated lymphoid tissues (NALT), cervical lymph nodes (CLN), mesenteric lymph nodes (MLN) and peripheral lymph nodes (PLN) using flow cytometry and in vitro proliferation assays. Overall, the NALT contained a higher proportion of APCs and a lower proportion of T cells compared to the CLN, MLN and PLN. The APCs of the NALT more often belonged to the CD11c+ CD11b+ and the CD11cneg CD11b+ subsets as compared to the other sites. Both of these APC populations showed little sign of activation, that is low expression of the markers CD40, CD86 and IAd. Instead, the APCs of the NALT more often co-expressed CX3CR1 and CD206, markers associated with a tolerogenic function. No increase in the proportion of regulatory T cells was observed in the NALT. Instead, the T cells frequently exhibited a memory/effector phenotype, expressing the homing markers α4β7, CCR4 and CCR9, but rarely the naïve phenotype cell surface marker CD45RB. In contrast, the T cells at the other sites were mostly of the naïve phenotype. In addition, cells from the NALT did not proliferate upon in vitro stimulation with Con A, whereas the cells from the other sites did. Taken together, these results suggest that the NALT is primarily an effector site rather than one for activation and differentiation, despite it being regarded as a site of induction.
Collapse
Affiliation(s)
- M Bankvall
- Department of Oral Medicine & Pathology, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - M Jontell
- Department of Oral Medicine & Pathology, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - A Wold
- Department of Infectious Diseases, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - S Östman
- Department of Infectious Diseases, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
7
|
Howe SE, Sowa G, Konjufca V. Systemic and Mucosal Antibody Responses to Soluble and Nanoparticle-Conjugated Antigens Administered Intranasally. Antibodies (Basel) 2016; 5:antib5040020. [PMID: 31558001 PMCID: PMC6698832 DOI: 10.3390/antib5040020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 07/16/2016] [Accepted: 09/18/2016] [Indexed: 02/03/2023] Open
Abstract
Nanoparticles (NPs) are increasingly being used for drug delivery, as well as antigen carriers and immunostimulants for the purpose of developing vaccines. In this work, we examined how intranasal (i.n.) priming followed by i.n. or subcutaneous (s.c.) boosting immunization affects the humoral immune response to chicken ovalbumin (Ova) and Ova conjugated to 20 nm NPs (NP-Ova). We show that i.n. priming with 20 mg of soluble Ova, a dose known to trigger oral tolerance when administered via gastric gavage, induced substantial systemic IgG1 and IgG2c, as well as mucosal antibodies. These responses were further boosted following a s.c. immunization with Ova and complete Freund’s adjuvant (Ova+CFA). In contrast, 100 µg of Ova delivered via NPs induced an IgG1-dominated systemic response, and primed the intestinal mucosa for secretion of IgA. Following a secondary s.c. or i.n. immunization with Ova+CFA or NP-Ova, systemic IgG1 titers significantly increased, and serum IgG2c and intestinal antibodies were induced in mice primed nasally with NP-Ova. Only Ova- and NP-Ova-primed mice that were s.c.-boosted exhibited substantial systemic and mucosal titers for up to 6 months after priming, whereas the antibodies of i.n.-boosted mice declined over time. Our results indicate that although the amount of Ova delivered by NPs was 1000-fold less than Ova delivered in soluble form, the antigen-specific antibody responses, both systemic and mucosal, are essentially identical by 6 months following the initial priming immunization. Additionally, both i.n.- and s.c.-boosting strategies for NP-Ova-primed mice were capable of inducing a polarized Th1/Th2 immune response, as well as intestinal antibodies; however, it is only by using a heterogeneous prime-boost strategy that long-lasting antibody responses were initiated. These results provide valuable insight for future mucosal vaccine development, as well as furthering our understanding of mucosal antibody responses.
Collapse
Affiliation(s)
- Savannah E Howe
- Department of Microbiology, Southern Illinois University, Carbondale, IL 62901, USA.
| | - Gavin Sowa
- Department of Chemistry, Southern Illinois University, Carbondale, IL 62901, USA.
| | - Vjollca Konjufca
- Department of Microbiology, Southern Illinois University, Carbondale, IL 62901, USA.
| |
Collapse
|
8
|
Stenner M, Rudack C. Diseases of the nose and paranasal sinuses in child. GMS CURRENT TOPICS IN OTORHINOLARYNGOLOGY, HEAD AND NECK SURGERY 2014; 13:Doc10. [PMID: 25587370 PMCID: PMC4273171 DOI: 10.3205/cto000113] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Diseases of the pediatric nose and nasal sinuses as well as neighboring anatomical structures encompass a variety of pathologies, especially of inflammatory nature. Congenital disease, such as malformations and structural deviations of the nasal septum, as well as systemic metabolic pathologies affecting the nose and sinuses, rarely require medical therapy from an Otolaryngologist. The immunological function of the mucosa and genetic factors play a role in the development of disease in the pediatric upper airway tract, especially due to the constantly changing anatomy in this growth phase. Disease description of the nose and nasal sinuses due to mid-facial growth must also take developmental age differences (infant, toddler, preschool, and school age) into account. Epidemiological examinations and evidence based studies are often lacking in the pediatric population. The wide range of inflammatory diseases of the nose and paranasal sinuses, such as the acute and chronic rhinosinusitis, the allergic rhinitis, and adenoid disease, play a role in the susceptibility of a child to infection. The susceptibility to infection depends on the pediatric age structure (infant, young child) and has yet to be well defined. The acute rhinosinusitis in children develops after a viral infection of the upper airways, also referred to as the "common cold" in the literature. It usually spontaneously heals within ten days without any medical therapy. Antibiotic therapy is prudent in complicated episodes of ARS. The antibiotic therapy is reserved for children with complications or associated disease, such as bronchial asthma and/or chronic bronchitis. A chronic rhinosinusitis is defined as the inflammatory change in the nasal mucosa and nasal sinus mucosa, in which the corresponding symptoms persist for over 12 weeks. The indication for CT-imaging of the nasal sinuses is reserved for cases of chronic rhinosinusitis that have been successfully treated with medication. A staged therapeutic concept is followed in CRS based on conservative and surgical methods. Nasal sinus surgery is considered nowadays as effective and safe in children. Based on the assumption that adenoids are a reservoir for bacteria, from which recurrent infections of the nose and nasal sinus originate, the adenoidectomy is still defined as a cleansing procedure in rhinosinusitis. 69.3% of the children had benefit from adenoidectomy. Comorbidities, such as pediatric bronchial asthma, presently play an even more important role in the therapy of rhinosinusitis; therefore, it is often wise to have the support of pediatricians. In western European countries 40% of children presently suffer from allergic rhinitis, in which pronounced nasal obstruction can cause disturbed growth in facial bones. An early therapy with SIT may prevent the development of bronchial asthma and secondary sensitization to other allergens. Therefore, SIT is recommended in treatment of allergic rhinitis whenever, if possible. The assessment of diagnostic tools is for the examiner not often possible due to the lack of evidence. Rhinosurgical approaches are often described in study reports; however, they lack the standard prospective randomized long-term study design required nowadays and can only be evaluated with caution in the literature.
Collapse
Affiliation(s)
- Markus Stenner
- Klinik für Hals-, Nasen- und Ohrenheilkunde, Universitätsklinikum Münster, Germany
| | - Claudia Rudack
- Klinik für Hals-, Nasen- und Ohrenheilkunde, Universitätsklinikum Münster, Germany
| |
Collapse
|
9
|
Sands BE, Feagan BG, Rutgeerts P, Colombel JF, Sandborn WJ, Sy R, D'Haens G, Ben-Horin S, Xu J, Rosario M, Fox I, Parikh A, Milch C, Hanauer S. Effects of vedolizumab induction therapy for patients with Crohn's disease in whom tumor necrosis factor antagonist treatment failed. Gastroenterology 2014; 147:618-627.e3. [PMID: 24859203 DOI: 10.1053/j.gastro.2014.05.008] [Citation(s) in RCA: 524] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 04/10/2014] [Accepted: 05/19/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS There is an increasing need for new treatments for patients with Crohn's disease (CD) in whom previous therapy with tumor necrosis factor (TNF) antagonists has failed. We performed a placebo-controlled, phase 3, double-blind trial to evaluate the efficacy and safety of vedolizumab, an antibody against the integrin α4β7, as induction therapy. METHODS Patients with moderately to severely active CD (CD activity index [CDAI] score, 220-400 points) were assigned randomly to groups given vedolizumab (300 mg) or placebo intravenously at weeks 0, 2, and 6. The primary analysis involved 315 patients with previous TNF antagonist failure (ie, an inadequate response to, loss of response to, or intolerance of ≥1 TNF antagonists); we determined the proportion of patients in clinical remission (CDAI, ≤150 points) at week 6. Secondary analyses evaluated outcomes at weeks 6 and 10 in this population and in the overall population (N = 416), which included patients naive to TNF antagonist therapy (n = 101). RESULTS Among patients who had experienced previous TNF antagonist failure, 15.2% of those given vedolizumab and 12.1% of those given placebo were in remission at week 6 (P = .433). At week 10, a higher proportion of this population given vedolizumab was in remission (26.6%) than those given placebo (12.1%) (nominal P = .001; relative risk, 2.2; 95% confidence interval, 1.3-3.6). A higher proportion of patients with previous TNF antagonist failure given vedolizumab also had a CDAI-100 response (≥100-point decrease in CDAI score from baseline) at week 6 than those given placebo (39.2% vs 22.3%; nominal P = .001; relative risk, 1.8; 95% confidence interval, 1.2-2.5). Adverse event results were similar among all groups. CONCLUSIONS Vedolizumab was not more effective than placebo in inducing clinical remission at week 6 among patients with CD in whom previous treatment with TNF antagonists had failed. The therapeutic benefits of vedolizumab in these patients were detectable at week 10. ClinicalTrials.gov number: NCT01224171.
Collapse
Affiliation(s)
- Bruce E Sands
- The Dr Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Brian G Feagan
- Robarts Clinical Trials Inc, and Robarts Research Institute, Department of Medicine, and Department of Epidemiology and Biostatistics, University of Western Ontario, London, Ontario, Canada
| | - Paul Rutgeerts
- Department of Gastroenterology, University Hospital Gasthuisberg and Katholieke Universiteit, Leuven, Belgium
| | - Jean-Frédéric Colombel
- The Dr Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, New York; Département d'Hépato-Gastroentérologie et Centre d'Investigations Cliniques, Centre Hospitalier Régional Universitaire de Lille, Université Lille Nord de France, Lille, France
| | - William J Sandborn
- Division of Gastroenterology, University of California San Diego, La Jolla, California
| | - Richmond Sy
- Department of Gastroenterology, University of Ottawa, Ottawa, Ontario, Canada
| | - Geert D'Haens
- Department of Gastroenterology, Inflammatory Bowel Disease Centre of the Academic Medical Centre, Amsterdam, the Netherlands
| | - Shomron Ben-Horin
- Department of Gastroenterology, Sheba Medical Center and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jing Xu
- Takeda Pharmaceuticals International Co, Cambridge, Massachusetts
| | - Maria Rosario
- Takeda Pharmaceuticals International Co, Cambridge, Massachusetts
| | - Irving Fox
- Takeda Pharmaceuticals International Co, Cambridge, Massachusetts
| | - Asit Parikh
- Takeda Pharmaceuticals International, Inc, Deerfield, Illinois
| | - Catherine Milch
- Takeda Pharmaceuticals International Co, Cambridge, Massachusetts
| | - Stephen Hanauer
- Division of Gastroenterology, The University of Chicago Medical Center, Chicago, Illinois
| |
Collapse
|
10
|
Casteleyn C, Cornillie P, Van Ginneken C, Simoens P, Van Cruchten S, Vandevelde K, Van den Broeck W. Lymph drainage from the ovine tonsils: an anatomical study of the tonsillar lymph vessels. Anat Histol Embryol 2014; 43:482-9. [PMID: 24597835 DOI: 10.1111/ahe.12107] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 02/01/2014] [Indexed: 01/17/2023]
Abstract
Although the tonsils of sheep have gained much attention during the last decade, only few data are available on their lymph vessel architecture. Tonsillar lymph vessels are immunologically important as they form the efferent routes for locally activated immune cells to reach the draining lymph nodes. To gain insight into the tonsillar lymph drainage in the sheep, Indian ink and a casting polymer were injected into the interstitium of the five tonsils present in the heads of slaughtered sheep. This enabled us to determine the draining lymph node and to examine the microscopic organization of lymph vessels using light and scanning electron microscopy. No lymph vessels were observed within the tonsillar lymphoid follicles. The corrosion casts demonstrated that the lymphoid follicles are surrounded by numerous sacculated lymph sinuses that drain into a dense interfollicular lymph vessel network. From here, the lymph flows into single small lymph vessels that in turn drain into larger lymph vessels extending towards the medial retropharyngeal lymph node. The presented results can be valuable for immunological studies, for example during oral or intranasal vaccine development.
Collapse
Affiliation(s)
- C Casteleyn
- Department of Morphology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium; Laboratory of Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | | | | | | | | | | | | |
Collapse
|
11
|
Fu J, Liang J, Kang H, Lin J, Yu Q, Yang Q. Effects of different CpG oligodeoxynucleotides with inactivated avian H5N1 influenza virus on mucosal immunity of chickens. Poult Sci 2013; 92:2866-75. [PMID: 24135589 DOI: 10.3382/ps.2013-03205] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Oligodeoxynucleotide containing unmethylated CpG motifs (CpG-ODN) has been proved to be a potent and safe vaccine adjuvant. However, the application of CpG-ODN in poultry vaccines was limited because of its high cost to benefit ratio. The objective of this study was to identify the CpG-ODN with efficient adjuvant activity and low cost in chickens. Four sequences of CpG-ODN were designed based on CpG-ODN 2006, which was used as a template and positive sequence in our study. In the current study, in vitro observations revealed that the designed CpG-ODN had efficient immunostimulatory effects on chicken splenic lymphocytes. The in vivo results showed that the mRNA expressions of IL-6, IL-12, interferon-γ, and Toll-like receptor (TLR) 21 in upper respiratory tract tissues increased significantly in the early period after intranasal immunization with inactivated avian H5N1 influenza virus (IAIV) and CpG-ODN (P < 0.01). In addition, the avian influenza virus (AIV)-specific secretory IgA antibody level in the lavage fluid of upper respiratory tract increased significantly after intranasal immunization with IAIV and CpG-ODN, so did AIV-specific IgG in serum (P < 0.01). Among all the designed CpG-ODN, CpG-ODN F3 with an addition of poly-guanosine strings at the 3'-end not only had the best enhancement on local mucosal immune response but also showed an effective induction of systemic immune response. Most importantly, the virus challenge study showed that prior administration of IAIV with CpG-ODN F3 could protect chickens effectively against live AIV H5N1 challenge. Additionally, among all the CpG-ODN in our study, the cost of the designed CpG-ODN F3 was the lowest because of the partially phosphorothioate backbone. Therefore, we speculated that CpG-ODN F3 with efficient adjuvant activity and a big cost advantage over CpG-ODN F1 (CpG-ODN 2006) might serve as an efficient and affordable nasal adjuvant for inactivated AIV vaccine in chicken.
Collapse
Affiliation(s)
- Jia Fu
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, China
| | | | | | | | | | | |
Collapse
|
12
|
Pettini E, Prota G, Ciabattini A, Boianelli A, Fiorino F, Pozzi G, Vicino A, Medaglini D. Vaginal immunization to elicit primary T-cell activation and dissemination. PLoS One 2013; 8:e80545. [PMID: 24349003 PMCID: PMC3857820 DOI: 10.1371/journal.pone.0080545] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 10/14/2013] [Indexed: 01/30/2023] Open
Abstract
Primary T-cell activation at mucosal sites is of utmost importance for the development of vaccination strategies. T-cell priming after vaginal immunization, with ovalbumin and CpG oligodeoxynucleotide adjuvant as model vaccine formulation, was studied in vivo in hormone-synchronized mice and compared to the one induced by the nasal route. Twenty-four hours after both vaginal or nasal immunization, antigen-loaded dendritic cells were detected within the respective draining lymph nodes. Vaginal immunization elicited a strong recruitment of antigen-specific CD4(+) T cells into draining lymph nodes that was more rapid than the one observed following nasal immunization. T-cell clonal expansion was first detected in iliac lymph nodes, draining the genital tract, and proliferated T cells disseminated towards distal lymph nodes and spleen similarly to what observed following nasal immunization. T cells were indeed activated by the antigen encounter and acquired homing molecules essential to disseminate towards distal lymphoid organs as confirmed by the modulation of CD45RB, CD69, CD44 and CD62L marker expression. A multi-type Galton Watson branching process, previously used for in vitro analysis of T-cell proliferation, was applied to model in vivo CFSE proliferation data in draining lymph nodes 57 hours following immunization, in order to calculate the probabilistic decision of a cell to enter in division, rest in quiescence or migrate/die. The modelling analysis indicated that the probability of a cell to proliferate was higher following vaginal than nasal immunization. All together these data show that vaginal immunization, despite the absence of an organized mucosal associated inductive site in the genital tract, is very efficient in priming antigen-specific CD4(+) T cells and inducing their dissemination from draining lymph nodes towards distal lymphoid organs.
Collapse
Affiliation(s)
- Elena Pettini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Gennaro Prota
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Annalisa Ciabattini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Alessandro Boianelli
- Dipartimento di Ingegneria dell'Informazione (DII), Centro per lo Studio dei Sistemi Complessi (CSC), Università di Siena, Siena, Italy
| | - Fabio Fiorino
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Gianni Pozzi
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Antonio Vicino
- Dipartimento di Ingegneria dell'Informazione (DII), Centro per lo Studio dei Sistemi Complessi (CSC), Università di Siena, Siena, Italy
| | - Donata Medaglini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
- * E-mail:
| |
Collapse
|
13
|
Fiorino F, Pettini E, Pozzi G, Medaglini D, Ciabattini A. Prime-boost strategies in mucosal immunization affect local IgA production and the type of th response. Front Immunol 2013; 4:128. [PMID: 23755051 PMCID: PMC3665932 DOI: 10.3389/fimmu.2013.00128] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 05/15/2013] [Indexed: 12/21/2022] Open
Abstract
Combinations of different delivery routes for priming and boosting represent vaccination strategies that can modulate magnitude, quality, and localization of the immune response. A murine model was used to study T cell clonal expansion following intranasal (IN) or subcutaneous (SC) priming, and secondary immune responses after boosting by either homologous or heterologous routes. T cell primary activation was studied by using the adoptive transfer model of ovalbumin-specific transgenic CD4(+) T cells. Both IN and SC immunization efficiently elicited, in the respective draining lymph nodes, primary clonal expansion of antigen-specific CD4(+) T cells that disseminated toward distal lymph nodes (mesenteric and iliac) and the spleen. After boosting, a significant serum IgG response was induced in all groups independent of the combination of immunization routes used, while significant levels of local IgA were detected only in mice boosted by the IN route. Mucosal priming drove a stronger Th1 polarization than the systemic route, as shown by serum IgG subclass analysis. IFN-gamma production was observed in splenocytes of all groups, while prime-boost vaccine combinations that included the mucosal route, yielded higher levels of IL-17. Memory lymphocytes were identified in both spleen and draining lymph nodes in all immunized mice, with the highest number of IL-2 producing cells detected in mice primed and boosted by the nasal route. This work shows the critical role of immunization routes in modulating quality and localization of immune responses in prime-boost vaccine strategies.
Collapse
Affiliation(s)
- Fabio Fiorino
- Laboratorio di Microbiologia Molecolare e Biotecnologia, Dipartimento di Biotecnologie Mediche, Università di Siena , Siena , Italy
| | | | | | | | | |
Collapse
|
14
|
Zbrun M, Zielinski G, Piscitelli H, Descarga C, Urbani L, Defain Tesoriero M, Hermida L. Evaluation of anti-Moraxella bovis pili immunoglobulin-A in tears following intranasal vaccination of cattle. Res Vet Sci 2012; 93:183-9. [DOI: 10.1016/j.rvsc.2011.05.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 05/05/2011] [Accepted: 05/12/2011] [Indexed: 11/30/2022]
|
15
|
Kang H, Wang H, Yu Q, Yang Q. Effect of intranasal immunization with inactivated avian influenza virus on local and systemic immune responses in ducks. Poult Sci 2012; 91:1074-80. [DOI: 10.3382/ps.2011-01817] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
16
|
The tonsils revisited: review of the anatomical localization and histological characteristics of the tonsils of domestic and laboratory animals. Clin Dev Immunol 2011; 2011:472460. [PMID: 21869895 PMCID: PMC3159307 DOI: 10.1155/2011/472460] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Accepted: 06/09/2011] [Indexed: 01/08/2023]
Abstract
This paper gives an overview of the anatomical localization and histological characteristics of the tonsils that are present in ten conventional domestic animal species, including the sheep, goat, ox, pig, horse, dog, cat, rabbit, rat, and pigeon. Anatomical macrographs and histological images of the tonsils are shown. Six tonsils can be present in domestic animals, that is, the lingual, palatine, paraepiglottic, pharyngeal, and tubal tonsils and the tonsil of the soft palate. Only in the sheep and goat, all six tonsils are present. Proper tonsils are absent in the rat, and pigeon. In the rabbit, only the palatine tonsils can be noticed, whereas the pig does not present palatine tonsils. The paraepiglottic tonsils lack in the ox, horse, and dog. In addition, the dog and cat are devoid of the tubal tonsil and the tonsil of the soft palate.
Collapse
|
17
|
Novotny LA, Clements JD, Bakaletz LO. Transcutaneous immunization as preventative and therapeutic regimens to protect against experimental otitis media due to nontypeable Haemophilus influenzae. Mucosal Immunol 2011; 4:456-67. [PMID: 21326197 PMCID: PMC3118858 DOI: 10.1038/mi.2011.6] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Accepted: 01/18/2011] [Indexed: 02/04/2023]
Abstract
We have developed three nontypeable Haemophilus influenzae (NTHI) adhesin-derived immunogens that are significantly efficacious against experimental otitis media (OM) due to NTHI when delivered parenterally. We now expanded our preventative immunization strategies to include transcutaneous immunization (TCI) as a less invasive, but potentially equally efficacious, regimen to prevent OM due to NTHI. Additionally, we examined the potential of TCI as a therapeutic immunization regimen to resolve ongoing experimental OM. Preventative immunization with NTHI outer membrane protein (OMP) P5- and type IV pilus-targeted immunogens, delivered with the adjuvant LT(R192G-L211A), induced significantly earlier clearance of NTHI from the nasopharynges and middle ears of challenged chinchillas compared with receipt of immunogen or adjuvant alone. Moreover, therapeutic immunization resulted in significant resolution of established NTHI biofilms from the middle ear space of animals compared with controls. These data advocate TCI with the adhesin-directed immunogens as an efficacious regimen for prevention and resolution of experimental NTHI-induced OM.
Collapse
Affiliation(s)
- L A Novotny
- The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, Center for Microbial Pathogenesis and The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - J D Clements
- Department of Microbiology and Immunology, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
| | - L O Bakaletz
- The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, Center for Microbial Pathogenesis and The Ohio State University College of Medicine, Columbus, Ohio, USA
- Nationwide Children's Hospital, Department of Otolaryngology and The Ohio State University College of Medicine, Columbus, Ohio, USA
| |
Collapse
|
18
|
Batista A, Quattrocchi V, Olivera V, Langellotti C, Pappalardo J, Di Giacomo S, Mongini C, Portuondo D, Zamorano P. Adjuvant effect of Cliptox™ on the protective immune response induced by an inactivated vaccine against foot and mouth disease virus in mice. Vaccine 2010; 28:6361-6. [DOI: 10.1016/j.vaccine.2010.06.098] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Revised: 06/16/2010] [Accepted: 06/29/2010] [Indexed: 10/19/2022]
|
19
|
Rodriguez-Monroy MA, Moreno-Fierros L. Striking activation of NALT and nasal passages lymphocytes induced by intranasal immunization with Cry1Ac protoxin. Scand J Immunol 2010; 71:159-68. [PMID: 20415781 DOI: 10.1111/j.1365-3083.2009.02358.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cry1Ac protoxin from Bacillus thuringiensis is a potent mucosal immunogen and adjuvant. When delivered intranasally (i.n.) Cry1Ac elicits significant antibody response and is able to improve vaccination against Naegleria fowleri infection, but the functional effects occurring in nasal lymphocytes when this protein is administered alone have not been determined. Here, we investigated the effects of i.n. immunization with Cry1Ac on antibody production, lymphocyte activation and cytokine production in lymphocytes from nasal-associated lymphoid tissue (NALT) and nasal passages (NP). Our results show that i.n. immunization with Cry1Ac induced significant specific IgA and IgG cell responses, especially in NP. Besides, it increased the proportion of lymphocytes expressing the activation markers CD25 and CD69 in both nasal tissues, but differently. CD25 was increased in B cells along with CD4 and CD8 T cells from NALT and NP, while CD69 was increased in B cells from both tissues but only in CD4 T cells from NP. Finally, we found that Cry1Ac augmented especially a Th2 profile of cytokines, as the proportion of T cells that spontaneously produced IL-4, IL-5 and IL-10 was increased and this effect was higher in NP than in NALT. These data contribute to explain the potent immunogenicity of Cry1Ac via i.n. route.
Collapse
Affiliation(s)
- M A Rodriguez-Monroy
- Carrera de Médico Cirujano, Unidad de Biomedicina F.E.S. Iztacala, Universidad Nacional Autónoma de México, Tlalnepantia, Edo. México, México
| | | |
Collapse
|
20
|
NALT (nasal cavity-associated lymphoid tissue) in the rabbit. Vet Immunol Immunopathol 2010; 133:212-8. [DOI: 10.1016/j.vetimm.2009.08.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2009] [Revised: 08/06/2009] [Accepted: 08/10/2009] [Indexed: 11/19/2022]
|
21
|
Ciabattini A, Pettini E, Arsenijevic S, Pozzi G, Medaglini D. Intranasal immunization with vaccine vector Streptococcus gordonii elicits primed CD4+ and CD8+ T cells in the genital and intestinal tracts. Vaccine 2009; 28:1226-33. [PMID: 19945415 DOI: 10.1016/j.vaccine.2009.11.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 10/30/2009] [Accepted: 11/09/2009] [Indexed: 01/01/2023]
Abstract
Generation of primed T cells is crucial for the development of optimal vaccination strategies. Using a TCR-transgenic CD4(+) and CD8(+) T cell adoptive transfer model, we demonstrate that a single nasal immunization with recombinant Streptococcus gordonii induces antigen-specific primed T cells in lymph nodes draining the genital and intestinal tracts with about 80% of CD4(+) and 50% of CD8(+) proliferating cells. T cell clonal expansion was also observed in cervical lymph nodes, draining the immunization site, and in the spleen. The modulation of CD44 and CD45RB marker expression indicated that proliferating T cells were activated. Proliferation in distal mesenteric and iliac lymph nodes and in the spleen was observed 5 days after nasal immunization, while in draining cervical lymph nodes proliferation peaked already at day 3. The division profile of transgenic T cells observed in iliac and mesenteric lymph nodes was discontinuous, showing the lack of early cell divisions. The kinetics of T cell clonal expansion, the discontinuous division profile and the modulation of migration markers such as CD62L suggest that activated antigen-specific T cells disseminate from the immunization site to distal intestinal and genital tracts. These data demonstrate the efficacy of nasal immunization with recombinant S. gordonii in eliciting CD4(+) and CD8(+) T cell priming not only in draining sites, but also in the genital and intestinal tracts and in the spleen.
Collapse
Affiliation(s)
- Annalisa Ciabattini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biologia Molecolare, Università di Siena, 53100 Siena, Italy
| | | | | | | | | |
Collapse
|
22
|
Xiaowen Z, Qinghua Y, Xiaofei Z, Qian Y. Co-administration of inactivated avian influenza virus with CpG or rIL-2 strongly enhances the local immune response after intranasal immunization in chicken. Vaccine 2009; 27:5628-32. [PMID: 19647063 DOI: 10.1016/j.vaccine.2009.07.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2007] [Revised: 07/10/2009] [Accepted: 07/10/2009] [Indexed: 11/15/2022]
Abstract
Intranasal delivery of vaccines is the most effective means of inducing effective immunity in the upper respiratory tract as well as other mucosal lymphoid tissues. To evaluate the effects of the H5N2 inactivated virus with adjuvant, 120 one-day-old chicks were intranasal immunized with the H5N2 inactivated virus respectively mixed with adjuvant CpG or recombinant IL-2 (rIL-2). The local immunocompetent cells on the respiratory tract were detected. The results showed that the number of intraepithelial lymphocytes (IELs), CD3(+) T lymphocytes and mast cells in respiratory tract increased significantly respectively and the number of IgA and IgG secreting cells increased significantly after immunization. However, there was no significant change in the immunocompetent cells of the animals administrated H5N2 inactivated virus alone compared to the control group. Our results indicated that intranasal administration of H5N2 inactivated virus with adjuvant CpG or rIL-2 could be beneficial to the local immune response in the respiratory tract.
Collapse
Affiliation(s)
- Zhang Xiaowen
- Key laboratory of animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China
| | | | | | | |
Collapse
|
23
|
Adair BM. Nanoparticle vaccines against respiratory viruses. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2009; 1:405-14. [PMID: 20049806 PMCID: PMC7169756 DOI: 10.1002/wnan.45] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Influenza virus, respiratory syncytial virus (RSV), and parainfluenza type 3 virus (PI-3V) are the major viral agents which are consistently involved in causing lower respiratory tract disease in humans and animals. The virus infection begins in the upper respiratory tract, where immune responses are initiated, and then progresses to the lower respiratory tract where destruction of cells and tissues leads to bronchitis, bronchiolitis, and pneumonia, which is occasionally fatal. Nanoparticle vaccines, incorporating antigenic components from influenza, RSV, or PI-3V have been shown to be capable of stimulating mucosal and systemic immune responses, which can prevent the spread of infection to the lower respiratory tract. The encapsulation of viral proteins within nanoparticles may also facilitate production of respiratory vaccines which are efficacious in infants, where presence of maternally derived antibodies can reduce vaccine efficacy.
Collapse
Affiliation(s)
- Brian M Adair
- Virology Department, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Stormont, Belfast, Northern Ireland, UK.
| |
Collapse
|
24
|
Oral live vaccine strain-induced protective immunity against pulmonary Francisella tularensis challenge is mediated by CD4+ T cells and antibodies, including immunoglobulin A. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2009; 16:444-52. [PMID: 19211773 DOI: 10.1128/cvi.00405-08] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Francisella tularensis is an intracellular gram-negative bacterium and the etiological agent of pulmonary tularemia. Given the high degrees of infectivity in the host and of dissemination of bacteria following respiratory infection, immunization strategies that target mucosal surfaces are critical for the development of effective vaccines against this organism. In this study, we have characterized the efficacy of protective immunity against pneumonic tularemia following oral vaccination with F. tularensis LVS (live vaccine strain). Mice vaccinated orally with LVS displayed colocalization of LVS with intestinal M cells, with subsequent enhanced production of splenic antigen-specific gamma interferon and of systemic and mucosal antibodies, including immunoglobulin A (IgA). LVS-vaccinated BALB/c mice were highly protected against intranasal (i.n.) SCHU S4 challenge and exhibited significantly less bacterial replication in the lungs, liver, and spleen than mock-immunized animals. Depletion of CD4(+) T cells significantly abrogated the protective immunity, and mice deficient in B cells or IgA displayed partial protection against SCHU S4 challenge. These results suggest that oral vaccination with LVS induces protective immunity against i.n. challenge with F. tularensis SCHU S4 by a process mediated cooperatively by CD4(+) T cells and antibodies, including IgA.
Collapse
|
25
|
Primary activation of antigen-specific naive CD4+ and CD8+ T cells following intranasal vaccination with recombinant bacteria. Infect Immun 2008; 76:5817-25. [PMID: 18838521 DOI: 10.1128/iai.00793-08] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The primary activation of T-helper and T-cytotoxic cells following mucosal immunization with recombinant Streptococcus gordonii was studied in vivo by adoptive transfer of ovalbumin (OVA)-specific transgenic CD8(+) (OT-I) and CD4(+) (OT-II) T cells. A recombinant strain, expressing on the surface the vaccine antigen Ag85B-ESAT-6 from Mycobacterium tuberculosis fused to OVA T-helper and T-cytotoxic epitopes (peptides 323 to 339 and 257 to 264), was constructed and used to immunize C57BL/6 mice by the intranasal route. Recombinant, but not wild-type, bacteria induced OVA-specific CD4(+) and CD8(+) T-cell clonal expansion in cervical lymph nodes, lung, and spleen. OVA-specific CD4(+) and CD8(+) T-cell proliferation appeared first in cervical lymph nodes and later in the spleen, suggesting a possible migration of activated cells from the inductive site to the systemic district. A significant correlation between the percentages of CD4(+) and CD8(+) proliferating T cells was observed for each animal. The expression of CD69, CD44, and CD45RB on proliferating T lymphocytes changed as a function of the cell division number, confirming T-cell activation following the antigen encounter. These data indicate that intranasal immunization with recombinant S. gordonii is capable of inducing primary activation of naive antigen-specific CD4(+) and CD8(+) T cells, both locally and systemically.
Collapse
|
26
|
Intranasal immunization of mice with recombinant Streptococcus gordonii expressing NadA of Neisseria meningitidis induces systemic bactericidal antibodies and local IgA. Vaccine 2008; 26:4244-50. [PMID: 18582996 DOI: 10.1016/j.vaccine.2008.05.049] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2008] [Revised: 05/14/2008] [Accepted: 05/20/2008] [Indexed: 11/20/2022]
Abstract
NadA and NhhA, two surface proteins of serogroup B Neisseria meningitidis identified as candidate vaccine antigens, were expressed on the surface of the human oral commensal bacterium Streptococcus gordonii. Recombinant strains were used to immunize BALB/c mice by the intranasal route and the local and systemic immune response was assessed. Mice were inoculated with recombinant bacteria administered alone or with LTR72, a partially inactivated mutant of Escherichia coli heat-labile enterotoxin, as a mucosal adjuvant. Intranasal immunization with live bacteria expressing NadA induced a significant serum antibody response, with a prevalence of the IgG2a subclass, bactericidal activity in the sera of 71% of animals, and a NadA-specific IgA response in nasal and bronchoalveolar lavages. A formalin-inactivated recombinant strain of S. gordonii expressing NadA was also administered intranasally, inducing a systemic and mucosal humoral response comparable to that of live bacteria. The administration of recombinant bacteria with the mucosal adjuvant LTR72 stimulated a stronger systemic antibody response, protective in 85% of sera, while did not increase the local IgA response. Recombinant S. gordonii expressing NhhA induced a systemic but not mucosal antibody response. These data support the role of NadA as vaccine candidate against serogroup B meningococci, and the use of S. gordonii as vector for intranasal vaccination.
Collapse
|
27
|
Rosas C, Van de Walle GR, Metzger SM, Hoelzer K, Dubovi EJ, Kim SG, Parrish CR, Osterrieder N. Evaluation of a vectored equine herpesvirus type 1 (EHV-1) vaccine expressing H3 haemagglutinin in the protection of dogs against canine influenza. Vaccine 2008; 26:2335-43. [PMID: 18407383 DOI: 10.1016/j.vaccine.2008.02.064] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Revised: 01/24/2008] [Accepted: 02/28/2008] [Indexed: 12/20/2022]
Abstract
In 2004, canine influenza virus (CIV) was identified as a respiratory pathogen of dogs for the first time and found to be closely related to H3N8 equine influenza virus (EIV). We generated a recombinant vectored vaccine that expresses H3 of a recent isolate of EIV using equine herpesvirus type 1 (EHV-1) as the delivery vehicle. This EHV-1 vectored vaccine exhibited robust and stable EIV H3 expression and induced a strong influenza virus-specific response in both mice and dogs upon intranasal or subcutaneous administration. Furthermore, upon challenge with the recent CIV isolate A/canine/PA/10915-07, protection of vaccinated dogs could be demonstrated by a significant reduction in clinical sings, and, more importantly, by a significant reduction in virus shedding. We concluded that the EHV-1/H3 recombinant vector can be a valuable alternative for protection of dogs against clinical disease induced by CIV and can significantly reduce virus spread.
Collapse
Affiliation(s)
- Cristina Rosas
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Sabirov A, Metzger DW. Mouse models for the study of mucosal vaccination against otitis media. Vaccine 2008; 26:1501-24. [PMID: 18295938 PMCID: PMC2323831 DOI: 10.1016/j.vaccine.2008.01.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Revised: 01/10/2008] [Accepted: 01/11/2008] [Indexed: 12/20/2022]
Abstract
Otitis media (OM) is one of the most common infectious diseases in humans. The pathogenesis of OM involves nasopharyngeal (NP) colonization and retrograde ascension of the pathogen up the Eustachian tube into the middle ear (ME). Due to increasing rates of antibiotic resistance, there is an urgent need for vaccines to prevent infections caused by the most common causes of bacterial OM, including nontypeable Haemophilus influenzae, Streptococcus pneumoniae and Moraxella catarrhalis. Current vaccine strategies aim to diminish bacterial NP carriage, thereby reducing the likelihood of developing acute OM. To be effective, vaccination should induce local mucosal immunity both in the ME and in the NP. Studies in animal models have demonstrated that the intranasal route of vaccination is particularly effective at inducing immune responses in the nasal passage and ME for protection against OM. The mouse is increasingly used in these models, because of the availability of murine reagents and the existence of technology to manipulate murine models of disease immunologically and genetically. Previous studies confirmed the suitability of the mouse as a model for inflammatory processes in acute OM. Here, we discuss various murine models of OM and review the applicability of these models to assess the efficacy of mucosal vaccination and the mechanisms responsible for protection. In addition, we discuss various mucosal vaccine antigens, mucosal adjuvants and mucosal delivery systems.
Collapse
Affiliation(s)
- Albert Sabirov
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York 12208
| | - Dennis W. Metzger
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York 12208
| |
Collapse
|
29
|
Bielinska AU, Janczak KW, Landers JJ, Makidon P, Sower LE, Peterson JW, Baker JR. Mucosal immunization with a novel nanoemulsion-based recombinant anthrax protective antigen vaccine protects against Bacillus anthracis spore challenge. Infect Immun 2007; 75:4020-9. [PMID: 17502384 PMCID: PMC1952013 DOI: 10.1128/iai.00070-07] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The currently available commercial human anthrax vaccine requires multiple injections for efficacy and has side effects due to its alum adjuvant. These factors limit its utility when immunizing exposed populations in emergent situations. We evaluated a novel mucosal adjuvant that consists of a nontoxic, water-in-oil nanoemulsion (NE). This material does not contain a proinflammatory component but penetrates mucosal surfaces to load antigens into dendritic cells. Mice and guinea pigs were intranasally immunized with recombinant Bacillus anthracis protective antigen (rPA) mixed in NE as an adjuvant. rPA-NE immunization was effective in inducing both serum anti-PA immunoglobulin G (IgG) and bronchial anti-PA IgA and IgG antibodies after either one or two mucosal administrations. Serum anti-PA IgG2a and IgG2b antibodies and PA-specific cytokine induction after immunization indicate a Th1-polarized immune response. rPA-NE immunization also produced high titers of lethal-toxin-neutralizing serum antibodies in both mice and guinea pigs. Guinea pigs nasally immunized with rPA-NE vaccine were protected against an intradermal challenge with approximately 1,000 times the 50% lethal dose ( approximately 1,000x LD(50)) of B. anthracis Ames strain spores (1.38 x 10(3) spores), which killed control animals within 96 h. Nasal immunization also resulted in 70% and 40% survival rates against intranasal challenge with 10x LD(50) and 100x LD(50) (1.2 x 10(6) and 1.2 x 10(7)) Ames strain spores. Our results indicate that NE can effectively adjuvant rPA for intranasal immunization. This potentially could lead to a needle-free anthrax vaccine requiring fewer doses and having fewer side effects than the currently available human vaccine.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Administration, Intranasal
- Animals
- Anthrax/prevention & control
- Anthrax Vaccines/immunology
- Antibodies, Bacterial/blood
- Antigens, Bacterial/immunology
- Antitoxins/blood
- Bacillus anthracis/immunology
- Bacterial Toxins/immunology
- Bronchoalveolar Lavage Fluid/immunology
- Disease Models, Animal
- Emulsions
- Female
- Guinea Pigs
- Humans
- Immunity, Mucosal
- Immunoglobulin G/blood
- Mice
- Mice, Inbred BALB C
- Mice, Inbred CBA
- Nanoparticles
- Survival Analysis
- Vaccination/methods
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Anna U Bielinska
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, 1150 West Medical Center Drive, Ann Arbor, MI 48109-0648, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Rodríguez-Monroy MA, Rojas-Hernández S, Moreno-Fierros L. Phenotypic and functional differences between lymphocytes from NALT and nasal passages of mice. Scand J Immunol 2007; 65:276-88. [PMID: 17309783 DOI: 10.1111/j.1365-3083.2006.01898.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Nasal-associated lymphoid tissue (NALT) and nasal passages (NP) are considered as inductive and effector sites, respectively. The differences among lymphocyte populations of these nasal compartments have not been clearly established. The aim of this work was to contribute to the characterization of NALT and NP lymphocytes in mice. We isolated lymphocytes from both compartments, determined the frequencies of B220(+) cells as well as CD8(+), CD4(+) T cells; and analysed the expression of CD69 and CD25. Besides we analysed the proportion of T cells producing IL-2, IL-4, IL-5, IL-10, IFN-gamma and TNF-alpha. We found differences between NALT and NP. Two populations of B cells, B220+(hi) and B220+(low) were clearly distinguished only in NP, but not in NALT. Both (hi) and (low) B220(+) cells expressed CD19, but only a fraction of the B220+(low) population, expressed the plasma cell marker CD138(+). More B than T lymphocytes, as well as higher frequencies of CD4(+) than CD8(+) T cells were found in both compartments. A small fraction of NK cells (CD3(-)DX5(+)) along with a significant proportion of double negative CD4(-)CD8(-)CD3(+)DX5(-) T cells was detected in both nasal tissues. Furthermore, as expected for a mucosal effector site, NP contained major proportions of B220(+), T CD4(+) and T CD8(+) cells expressing CD25 and CD69 in comparison to NALT. Likewise, the proportion of T cells spontaneously producing IL-2, IFN-gamma, and IL-4, was higher in NP than in NALT. These data provide further evidence indicating that distinctive phenotypic and functional features exist in the lymphocyte populations residing at NALT and NP.
Collapse
Affiliation(s)
- M A Rodríguez-Monroy
- Inmunidad en Mucosas UBIMED, FES-Iztacala, Universidad Nacional Autónoma de México, Los Reyes Iztacala, Tlalnepantla, Méx, México
| | | | | |
Collapse
|
31
|
Medaglini D, Ciabattini A, Cuppone AM, Costa C, Ricci S, Costalonga M, Pozzi G. In vivo activation of naive CD4+ T cells in nasal mucosa-associated lymphoid tissue following intranasal immunization with recombinant Streptococcus gordonii. Infect Immun 2006; 74:2760-6. [PMID: 16622213 PMCID: PMC1459748 DOI: 10.1128/iai.74.5.2760-2766.2006] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The antigen-specific primary activation of CD4+ T cells was studied in vivo by adoptive transfer of ovalbumin-specific transgenic T cells (KJ1-26+ CD4+) following intranasal immunization with recombinant Streptococcus gordonii. A strain of S. gordonii expressing on its surface a model vaccine antigen fused to the ovalbumin (OVA) peptide from position 323 to 339 was constructed and used to study the OVA-specific T-cell activation in nasal mucosa-associated lymphoid tissue (NALT), lymph nodes, and spleens of mice immunized by the intranasal route. The recombinant strain, but not the wild type, activated the OVA-specific CD4+ T-cell population in the NALT (89% of KJ1-26+ CD4+ T cells) just 3 days following immunization. In the cervical lymph nodes and in the spleen, the percentage of proliferating cells was initially low, but it reached the peak of activation at day 5 (90%). This antigen-specific clonal expansion of KJ1-26+ CD4+ T cells after intranasal immunization was obtained with live and inactivated recombinant bacteria, and it indicates that the NALT is the site of antigen-specific T-cell priming.
Collapse
Affiliation(s)
- Donata Medaglini
- LAMMB, Dipartimento di Biologia Molecolare, Università di Siena, Siena, Italy.
| | | | | | | | | | | | | |
Collapse
|
32
|
Rharbaoui F, Bruder D, Vidakovic M, Ebensen T, Buer J, Guzmán CA. Characterization of a B220+Lymphoid Cell Subpopulation with Immune Modulatory Functions in Nasal-Associated Lymphoid Tissues. THE JOURNAL OF IMMUNOLOGY 2005; 174:1317-24. [PMID: 15661888 DOI: 10.4049/jimmunol.174.3.1317] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Complex mechanisms operate on mucosal tissues to regulate immune responsiveness and tolerance. When the lymphocyte subpopulations from murine nasal-associated lymphoid tissues (NALT) were characterized, we observed an accumulation of B220(low)CD3(low)CD4(-)CD8(-)CD19(-)c-Kit(+) cells. TCR transgenic mice and athymic mice were used for monitoring T cell lineage and the presence of extrathymic T cell precursors. The majority of cells from NALT exhibited a T cell precursor phenotype (CD4(-)CD8(-)CD19(-)c-Kit(+)). Fas-independent apoptosis was their main mechanism of cell death. We also demonstrated that B220(low)CD4(-)CD8(-)CD19(-) cells from NALT exhibited the potential to down-regulate the activation of mature T cells. However, the innate immunity receptor TLR2 was also highly expressed by this cell subpopulation. Moreover, nasal stimulation with a TLR2/6 agonist resulted in a partial activation of the double-negative cells. These results suggest that the immune responses in NALT may be in part modulated by a cell subpopulation that maintains a tolerogenic milieu by its proapoptotic status and suppressive activity, which can be reverted through stimulation of a TLR signaling cascade.
Collapse
Affiliation(s)
- Faiza Rharbaoui
- Division of Microbiology, GBF-German Research Center for Biotechnology, Braunschweig, Germany
| | | | | | | | | | | |
Collapse
|