Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
2.
Immunity ; 55(11): 2118-2134.e6, 2022 Nov 08.
Article in English | MEDLINE | ID: covidwho-2286532

ABSTRACT

While blood antibodies mediate protective immunity in most organs, whether they protect nasal surfaces in the upper airway is unclear. Using multiple viral infection models in mice, we found that blood-borne antibodies could not defend the olfactory epithelium. Despite high serum antibody titers, pathogens infected nasal turbinates, and neurotropic microbes invaded the brain. Using passive antibody transfers and parabiosis, we identified a restrictive blood-endothelial barrier that excluded circulating antibodies from the olfactory mucosa. Plasma cell depletions demonstrated that plasma cells must reside within olfactory tissue to achieve sterilizing immunity. Antibody blockade and genetically deficient models revealed that this local immunity required CD4+ T cells and CXCR3. Many vaccine adjuvants failed to generate olfactory plasma cells, but mucosal immunizations established humoral protection of the olfactory surface. Our identification of a blood-olfactory barrier and the requirement for tissue-derived antibody has implications for vaccinology, respiratory and CNS pathogen transmission, and B cell fate decisions.


Subject(s)
B-Lymphocytes , Plasma Cells , Animals , Mice , T-Lymphocytes , Immunoglobulins , Brain , Immunity, Mucosal , Antibodies, Viral
3.
Biomedicines ; 10(10)2022 Sep 29.
Article in English | MEDLINE | ID: covidwho-2065695

ABSTRACT

The waning effectiveness of the primary vaccination for SARS-CoV-2 led to administration of an additional booster dose (BD). The efficacy of the BD in stimulating humoral systemic immune response is well established, but its effectiveness on inducing mucosal immune reaction has not yet been reported. To address this issue, we evaluated SARS-CoV-2-specific antibody responses in the serum, saliva, and tears after BNT162b2 (Pfizer/BioNTech, New York, NY, USA) vaccination and BD, as well as after SARS-CoV-2 infection. After two doses of BNT162b2 vaccine, we observed specific serum IgG in 100% and IgA in 97.2% of subjects, associated with mucosal response in both salivary samples (sIgA in 97.2% and IgG(S) in 58.8%) and in tears (sIgA in 77.8% and IgG(S) in 67.7%). BD induced a recovery of the systemic humoral response and of tear sIgA when compared to 6 months of follow-up titers (p < 0.001; p = 0.012). However, sIgA levels in both tears and saliva were significantly lower following BD when compared to patients with prior SARS-CoV-2 infection (p = 0.001 and p = 0.005, respectively). Our results demonstrated that administration of BD restored high serum levels of both IgG and IgA but had a poor effect in stimulating mucosal immunity when compared to prior SARS-CoV-2 infection.

4.
Immunity ; 55(5): 800-818, 2022 05 10.
Article in English | MEDLINE | ID: covidwho-1838896

ABSTRACT

Pathogenic enteric viruses are a major cause of morbidity and mortality, particularly among children in developing countries. The host response to enteric viruses occurs primarily within the mucosa, where the intestinal immune system must balance protection against pathogens with tissue protection and tolerance to harmless commensal bacteria and food. Here, we summarize current knowledge in natural immunity to enteric viruses, highlighting specialized features of the intestinal immune system. We further discuss how knowledge of intestinal anti-viral mechanisms can be translated into vaccine development with particular focus on immunization in the oral route. Research reveals that the intestine is a complex interface between enteric viruses and the host where environmental factors influence susceptibility and immunity to infection, while viral infections can have lasting implications for host health. A deeper mechanistic understanding of enteric anti-viral immunity with this broader context can ultimately lead to better vaccines for existing and emerging viruses.


Subject(s)
Enterovirus Infections , Vaccines , Viruses , Antigens, Viral , Child , Humans , Immunity, Innate , Intestinal Mucosa , Intestines
5.
BioTech (Basel) ; 11(1)2022 Jan 30.
Article in English | MEDLINE | ID: covidwho-1760372

ABSTRACT

Infectious diseases caused by mucosal pathogens significantly increase mortality and morbidity. Thus, the possibility to target these pathogens at their primary entry points can consolidate protective immunity. Regarding SARS-CoV-2 infection, it has been observed that the upper respiratory mucosa is highly affected and that dysregulation of resident microbiota in the gut-lung axis plays a crucial role in determining symptom severity. Thus, understanding the possibility of eliciting various mucosal and adaptive immune responses allows us to effectively design bacterial mucosal vaccine vectors. Such design requires rationally selecting resident bacterial candidates as potential host carriers, evaluating effective carrier proteins for stimulating an immune response, and combining these two to improve antigenic display and immunogenicity. This review investigated mucosal vaccine vectors from 2015 to present, where a few have started to utilize Salmonella and lactic acid bacteria (LAB) to display SARS-CoV-2 Spike S proteins or fragments. Although current literature is still lacking for its studies beyond in vitro or in vivo efficiency, decades of research into these vectors show promising results. Here, we discuss the mucosal immune systems focusing on the gut-lung axis microbiome and offer new insight into the potential use of alpha streptococci in the upper respiratory tract as a vaccine carrier.

7.
Viruses ; 12(8)2020 07 27.
Article in English | MEDLINE | ID: covidwho-1512665

ABSTRACT

Acute viral bronchiolitis causes significant mortality in the developing world, is the number one cause of infant hospitalisation in the developed world, and is associated with the later development of chronic lung diseases such as asthma. A vaccine against respiratory syncytial virus (RSV), the leading cause of viral bronchiolitis in infancy, remains elusive, and hence new therapeutic modalities are needed to limit disease severity. However, much remains unknown about the underlying pathogenic mechanisms. Neutrophilic inflammation is the predominant phenotype observed in infants with both mild and severe disease, however, a clear understanding of the beneficial and deleterious effects of neutrophils is lacking. In this review, we describe the multifaceted roles of neutrophils in host defence and antiviral immunity, consider their contribution to bronchiolitis pathogenesis, and discuss whether new approaches that target neutrophil effector functions will be suitable for treating severe RSV bronchiolitis.


Subject(s)
Bronchiolitis, Viral/immunology , Bronchiolitis, Viral/pathology , Immunity, Innate , Neutrophils/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus, Human/immunology , Acute Disease , Animals , Clinical Trials as Topic , Humans , Inflammation/virology , Lung/virology , Mice , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus, Human/pathogenicity
8.
Immunology ; 160(2): 171-182, 2020 06.
Article in English | MEDLINE | ID: covidwho-11413

ABSTRACT

The airway epithelium represents a physical barrier to the external environment acting as the first line of defence against potentially harmful environmental stimuli including microbes and allergens. However, lung epithelial cells are increasingly recognized as active effectors of microbial defence, contributing to both innate and adaptive immune function in the lower respiratory tract. These cells express an ample repertoire of pattern recognition receptors with specificity for conserved microbial and host motifs. Modern molecular techniques have uncovered the complexity of the lower respiratory tract microbiome. The interaction between the microbiota and the airway epithelium is key to understanding how stable immune homeostasis is maintained. Loss of epithelial integrity following exposure to infection can result in the onset of inflammation in susceptible individuals and may culminate in lung disease. Here we discuss the current knowledge regarding the molecular and cellular mechanisms by which the pulmonary epithelium interacts with the lung microbiome in shaping immunity in the lung. Specifically, we focus on the interactions between the lung microbiome and the cells of the conducting airways in modulating immune cell regulation, and how defects in barrier structure and function may culminate in lung disease. Understanding these interactions is fundamental in the search for more effective therapies for respiratory diseases.


Subject(s)
Epithelial Cells/immunology , Lung Diseases/immunology , Lung/immunology , Microbiota/immunology , Respiratory Mucosa/immunology , Adaptive Immunity , Airway Remodeling/immunology , Homeostasis/immunology , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Immunity, Mucosal , Lung/cytology , Lung/microbiology , Lung Diseases/microbiology , Respiratory Mucosa/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL