Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 5.017
Filtrer
1.
Front Immunol ; 15: 1419527, 2024.
Article de Anglais | MEDLINE | ID: mdl-39286244

RÉSUMÉ

Mucosal barrier tissues and their mucosal associated lymphoid tissues (MALT) are attractive targets for vaccines and immunotherapies due to their roles in both priming and regulating adaptive immune responses. The upper and lower respiratory mucosae, in particular, possess unique properties: a vast surface area responsible for frontline protection against inhaled pathogens but also simultaneous tight regulation of homeostasis against a continuous backdrop of non-pathogenic antigen exposure. Within the upper and lower respiratory tract, the nasal and bronchial associated lymphoid tissues (NALT and BALT, respectively) are key sites where antigen-specific immune responses are orchestrated against inhaled antigens, serving as critical training grounds for adaptive immunity. Many infectious diseases are transmitted via respiratory mucosal sites, highlighting the need for vaccines that can activate resident frontline immune protection in these tissues to block infection. While traditional parenteral vaccines that are injected tend to elicit weak immunity in mucosal tissues, mucosal vaccines (i.e., that are administered intranasally) are capable of eliciting both systemic and mucosal immunity in tandem by initiating immune responses in the MALT. In contrast, administering antigen to mucosal tissues in the absence of adjuvant or costimulatory signals can instead induce antigen-specific tolerance by exploiting regulatory mechanisms inherent to MALT, holding potential for mucosal immunotherapies to treat autoimmunity. Yet despite being well motivated by mucosal biology, development of both mucosal subunit vaccines and immunotherapies has historically been plagued by poor drug delivery across mucosal barriers, resulting in weak efficacy, short-lived responses, and to-date a lack of clinical translation. Development of engineering strategies that can overcome barriers to mucosal delivery are thus critical for translation of mucosal subunit vaccines and immunotherapies. This review covers engineering strategies to enhance mucosal uptake via active targeting and passive transport mechanisms, with a parallel focus on mechanisms of immune activation and regulation in the respiratory mucosa. By combining engineering strategies for enhanced mucosal delivery with a better understanding of immune mechanisms in the NALT and BALT, we hope to illustrate the potential of these mucosal sites as targets for immunomodulation.


Sujet(s)
Immunité muqueuse , Immunomodulation , Humains , Animaux , Muqueuse respiratoire/immunologie , Muqueuse respiratoire/métabolisme , Tissu lymphoïde/immunologie , Vaccins/immunologie , Muqueuse nasale/immunologie , Muqueuse nasale/métabolisme , Administration par voie nasale
2.
J Exp Med ; 221(10)2024 Oct 07.
Article de Anglais | MEDLINE | ID: mdl-39240335

RÉSUMÉ

Predicting the immunogenicity of candidate vaccines in humans remains a challenge. To address this issue, we developed a lymphoid organ-chip (LO chip) model based on a microfluidic chip seeded with human PBMC at high density within a 3D collagen matrix. Perfusion of the SARS-CoV-2 spike protein mimicked a vaccine boost by inducing a massive amplification of spike-specific memory B cells, plasmablast differentiation, and spike-specific antibody secretion. Features of lymphoid tissue, including the formation of activated CD4+ T cell/B cell clusters and the emigration of matured plasmablasts, were recapitulated in the LO chip. Importantly, myeloid cells were competent at capturing and expressing mRNA vectored by lipid nanoparticles, enabling the assessment of responses to mRNA vaccines. Comparison of on-chip responses to Wuhan monovalent and Wuhan/Omicron bivalent mRNA vaccine boosts showed equivalent induction of Omicron neutralizing antibodies, pointing at immune imprinting as reported in vivo. The LO chip thus represents a versatile platform suited to the preclinical evaluation of vaccine-boosting strategies.


Sujet(s)
Vaccins contre la COVID-19 , COVID-19 , Cellules B mémoire , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Vaccins à ARNm , Humains , Vaccins contre la COVID-19/immunologie , Vaccins à ARNm/immunologie , SARS-CoV-2/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , Cellules B mémoire/immunologie , COVID-19/prévention et contrôle , COVID-19/immunologie , Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , Tissu lymphoïde/immunologie , Laboratoires sur puces , Vaccins synthétiques/immunologie , ARN messager/génétique , ARN messager/immunologie , ARN messager/métabolisme , Lymphocytes B/immunologie , Lymphocytes T CD4+/immunologie , Liposomes , Nanoparticules
3.
Nephrology (Carlton) ; 29 Suppl 2: 34-36, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39327765

RÉSUMÉ

IgA nephropathy is a mucosally driven disease and new therapeutic approaches are specifically targeting the mucosal production of IgA in the hope that this will lead to a reduction in circulating IgA immune complexes and mesangial IgA deposition. In this lecture, I discuss the rationale for targeting the mucosal immune system of the gut and the existing data from clinical trials supporting such an approach as a disease modifying treatment for IgA nephropathy.


Sujet(s)
Glomérulonéphrite à dépôts d'IgA , Immunoglobuline A , Muqueuse intestinale , Glomérulonéphrite à dépôts d'IgA/immunologie , Glomérulonéphrite à dépôts d'IgA/traitement médicamenteux , Glomérulonéphrite à dépôts d'IgA/métabolisme , Humains , Muqueuse intestinale/immunologie , Muqueuse intestinale/métabolisme , Immunoglobuline A/immunologie , Immunoglobuline A/métabolisme , Immunité muqueuse , Animaux , Microbiome gastro-intestinal , Résultat thérapeutique , Tissu lymphoïde/immunologie , Tissu lymphoïde/métabolisme
4.
J Exp Med ; 221(9)2024 09 02.
Article de Anglais | MEDLINE | ID: mdl-39093311

RÉSUMÉ

Shortly after the emergence of newly formed human B cells from bone marrow as transitional cells, they diverge along two developmental pathways that can be distinguished by the level of IgM they express and migratory biases. Here, we propose that differential tissue homing of immature B cell subsets contributes to human lymphoid tissue structure and function.


Sujet(s)
Mouvement cellulaire , Tissu lymphoïde , Humains , Tissu lymphoïde/immunologie , Tissu lymphoïde/cytologie , Mouvement cellulaire/immunologie , Lymphocytes B/immunologie , Immunoglobuline M/métabolisme , Immunoglobuline M/immunologie , Sous-populations de lymphocytes B/immunologie , Précurseurs lymphoïdes B/immunologie , Précurseurs lymphoïdes B/cytologie , Différenciation cellulaire/immunologie
5.
Signal Transduct Target Ther ; 9(1): 225, 2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39198425

RÉSUMÉ

Tertiary lymphoid structures (TLSs) are defined as lymphoid aggregates formed in non-hematopoietic organs under pathological conditions. Similar to secondary lymphoid organs (SLOs), the formation of TLSs relies on the interaction between lymphoid tissue inducer (LTi) cells and lymphoid tissue organizer (LTo) cells, involving multiple cytokines. Heterogeneity is a distinguishing feature of TLSs, which may lead to differences in their functions. Growing evidence suggests that TLSs are associated with various diseases, such as cancers, autoimmune diseases, transplant rejection, chronic inflammation, infection, and even ageing. However, the detailed mechanisms behind these clinical associations are not yet fully understood. The mechanisms by which TLS maturation and localization affect immune function are also unclear. Therefore, it is necessary to enhance the understanding of TLS development and function at the cellular and molecular level, which may allow us to utilize them to improve the immune microenvironment. In this review, we delve into the composition, formation mechanism, associations with diseases, and potential therapeutic applications of TLSs. Furthermore, we discuss the therapeutic implications of TLSs, such as their role as markers of therapeutic response and prognosis. Finally, we summarize various methods for detecting and targeting TLSs. Overall, we provide a comprehensive understanding of TLSs and aim to develop more effective therapeutic strategies.


Sujet(s)
Maladies auto-immunes , Structures lymphoïdes tertiaires , Humains , Structures lymphoïdes tertiaires/immunologie , Structures lymphoïdes tertiaires/anatomopathologie , Structures lymphoïdes tertiaires/génétique , Maladies auto-immunes/immunologie , Maladies auto-immunes/génétique , Maladies auto-immunes/thérapie , Maladies auto-immunes/anatomopathologie , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/génétique , Tumeurs/anatomopathologie , Inflammation/immunologie , Inflammation/génétique , Inflammation/anatomopathologie , Tissu lymphoïde/immunologie , Tissu lymphoïde/anatomopathologie , Animaux , Cytokines/immunologie , Cytokines/génétique
6.
Nature ; 632(8025): 637-646, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39085603

RÉSUMÉ

Nasal vaccination elicits a humoral immune response that provides protection from airborne pathogens1, yet the origins and specific immune niches of antigen-specific IgA-secreting cells in the upper airways are unclear2. Here we define nasal glandular acinar structures and the turbinates as immunological niches that recruit IgA-secreting plasma cells from the nasal-associated lymphoid tissues (NALTs)3. Using intact organ imaging, we demonstrate that nasal vaccination induces B cell expansion in the subepithelial dome of the NALT, followed by invasion into commensal-bacteria-driven chronic germinal centres in a T cell-dependent manner. Initiation of the germinal centre response in the NALT requires pre-expansion of antigen-specific T cells, which interact with cognate B cells in interfollicular regions. NALT ablation and blockade of PSGL-1, which mediates interactions with endothelial cell selectins, demonstrated that NALT-derived IgA-expressing B cells home to the turbinate region through the circulation, where they are positioned primarily around glandular acinar structures. CCL28 expression was increased in the turbinates in response to vaccination and promoted homing of IgA+ B cells to this site. Thus, in response to nasal vaccination, the glandular acini and turbinates provide immunological niches that host NALT-derived IgA-secreting cells. These cellular events could be manipulated in vaccine design or in the treatment of upper airway allergic responses.


Sujet(s)
Immunoglobuline A , Tissu lymphoïde , Muqueuse nasale , Plasmocytes , Lymphocytes T , Cornets , Animaux , Femelle , Mâle , Souris , Bactéries/immunologie , Mouvement cellulaire , Chimiokines CC/immunologie , Chimiokines CC/métabolisme , Centre germinatif/immunologie , Centre germinatif/cytologie , Immunoglobuline A/immunologie , Immunoglobuline A/métabolisme , Tissu lymphoïde/immunologie , Tissu lymphoïde/cytologie , Souris de lignée C57BL , Muqueuse nasale/cytologie , Muqueuse nasale/immunologie , Plasmocytes/immunologie , Plasmocytes/cytologie , Plasmocytes/métabolisme , Lymphocytes T/immunologie , Lymphocytes T/cytologie , Lymphocytes T/métabolisme , Cornets/cytologie , Cornets/immunologie , Vaccination , Administration par voie nasale , Vaccins/immunologie , Symbiose
7.
PLoS One ; 19(7): e0292408, 2024.
Article de Anglais | MEDLINE | ID: mdl-38950025

RÉSUMÉ

Co-infections are a common reality but understanding how the immune system responds in this context is complex and can be unpredictable. Heligmosomoides bakeri (parasitic roundworm, previously Heligmosomoides polygyrus) and Toxoplasma gondii (protozoan parasite) are well studied organisms that stimulate a characteristic Th2 and Th1 response, respectively. Several studies have demonstrated reduced inflammatory cytokine responses in animals co-infected with such organisms. However, while general cytokine signatures have been examined, the impact of the different cytokine producing lymphocytes on parasite control/clearance is not fully understood. We investigated five different lymphocyte populations (NK, NKT, γδ T, CD4+ T and CD8+ T cells), five organs (small intestine, Peyer's patches, mesenteric lymph nodes, spleen and liver), and 4 cytokines (IFN©, IL-4, IL-10 and IL-13) at two different time points (days 5 and 10 post T. gondii infection). We found that co-infected animals had significantly higher mortality than either single infection. This was accompanied by transient and local changes in parasite loads and cytokine profiles. Despite the early changes in lymphocyte and cytokine profiles, severe intestinal pathology in co-infected mice likely contributed to early mortality due to significant damage by both parasites in the small intestine. Our work demonstrates the importance of taking a broad view during infection research, studying multiple cell types, organs/tissues and time points to link and/or uncouple immunological from pathological findings. Our results provide insights into how co-infection with parasites stimulating different arms of the immune system can lead to drastic changes in infection dynamics.


Sujet(s)
Co-infection , Cytokines , Nematospiroides dubius , Toxoplasma , Animaux , Co-infection/immunologie , Co-infection/parasitologie , Toxoplasma/immunologie , Souris , Cytokines/métabolisme , Nematospiroides dubius/immunologie , Infections à Strongylida/immunologie , Infections à Strongylida/parasitologie , Infections à Strongylida/mortalité , Toxoplasmose/immunologie , Toxoplasmose/mortalité , Toxoplasmose/complications , Femelle , Toxoplasmose animale/immunologie , Toxoplasmose animale/mortalité , Toxoplasmose animale/parasitologie , Rate/immunologie , Rate/anatomopathologie , Rate/parasitologie , Charge parasitaire , Tissu lymphoïde/immunologie , Tissu lymphoïde/anatomopathologie , Tissu lymphoïde/parasitologie
8.
Chem Soc Rev ; 53(15): 7657-7680, 2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-38958009

RÉSUMÉ

Nanomaterials exhibit significant potential for stimulating immune responses, offering both local and systemic modulation across a variety of diseases. The lymphoid organs, such as the spleen and lymph nodes, are home to various immune cells, including monocytes and dendritic cells, which contribute to both the progression and prevention/treatment of diseases. Consequently, many nanomaterial formulations are being rationally designed to target these organs and engage with specific cell types, thereby inducing therapeutic and protective effects. In this review, we explore crucial cellular interactions and processes involved in immune regulation and highlight innovative nano-based immunomodulatory approaches. We outline essential considerations in nanomaterial design with an emphasis on their impact on biological interactions, targeting capabilities, and treatment efficacy. Through selected examples, we illustrate the strategic targeting of therapeutically active nanomaterials to lymphoid organs and the subsequent immunomodulation for infection resistance, inflammation suppression, self-antigen tolerance, and cancer immunotherapy. Additionally, we address current challenges, discuss emerging topics, and share our outlook on future developments in the field.


Sujet(s)
Immunomodulation , Inflammation , Nanostructures , Tumeurs , Humains , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Nanostructures/composition chimique , Inflammation/traitement médicamenteux , Inflammation/immunologie , Immunomodulation/effets des médicaments et des substances chimiques , Animaux , Immunothérapie , Tissu lymphoïde/immunologie , Tissu lymphoïde/effets des médicaments et des substances chimiques
9.
Vet Immunol Immunopathol ; 274: 110785, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38861830

RÉSUMÉ

The pig is emerging as a physiologically relevant biomedical large animal model. Delineating the functional roles of porcine adaptive T-lymphocyte subsets in health and disease is of critical significance, which facilitates mechanistic understanding of antigen-specific immune memory responses. We identified a novel T-helper/memory lymphocyte subset in pigs and performed phenotypic and functional characterization of these cells under steady state and following vaccination and infection with swine influenza A virus (SwIAV). A novel subset of CD3+CD4lowCD8α+CD8ß+ memory T-helper cells was identified in the blood of healthy adult pigs under homeostatic conditions. To understand the possible functional role/s of these cells, we characterized the antigen-specific T cell memory responses by multi-color flow cytometry in pigs vaccinated with a whole inactivated SwIAV vaccine, formulated with a phytoglycogen nanoparticle/STING agonist (ADU-S100) adjuvant (NanoS100-SwIAV). As a control, a commercial SwIAV vaccine was included in a heterologous challenge infection trial. The frequencies of antigen-specific IL-17A and IFNγ secreting CD3+CD4lowCD8α+CD8ß+ memory T-helper cells were significantly increased in the lung draining tracheobronchial lymph nodes (TBLN) of intradermal, intramuscular and intranasal inoculated NanoS100-SwIAV vaccine and commercial vaccine administered animals. While the frequencies of antigen-specific, IFNγ secreting CD3+CD4lowCD8α+CD8ß+ memory T-helper cells were significantly enhanced in the blood of intranasal and intramuscular vaccinates. These observations suggest that the CD3+CD4lowCD8α+CD8ß+ T-helper/memory cells in pigs may have a protective and/or regulatory role/s in immune responses against SwIAV infection. These observations highlight the heterogeneity and plasticity of porcine CD4+ T-helper/memory cells in response to respiratory viral infection in pigs. Comprehensive systems immunology studies are needed to further decipher the cellular lineages and functional role/s of this porcine T helper/memory cell subset.


Sujet(s)
Vaccins antigrippaux , Infections à Orthomyxoviridae , Maladies des porcs , Animaux , Suidae/immunologie , Vaccins antigrippaux/immunologie , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/médecine vétérinaire , Infections à Orthomyxoviridae/prévention et contrôle , Maladies des porcs/immunologie , Maladies des porcs/virologie , Maladies des porcs/prévention et contrôle , Lymphocytes T auxiliaires/immunologie , Appareil respiratoire/immunologie , Appareil respiratoire/virologie , Tissu lymphoïde/immunologie , Mémoire immunologique , Cellules T mémoire/immunologie , Sous-populations de lymphocytes T/immunologie , Virus de la grippe A/immunologie , Vaccination/médecine vétérinaire
10.
Front Immunol ; 15: 1400739, 2024.
Article de Anglais | MEDLINE | ID: mdl-38863701

RÉSUMÉ

Known for their distinct antigen-sampling abilities, microfold cells, or M cells, have been well characterized in the gut and other mucosa including the lungs and nasal-associated lymphoid tissue (NALT). More recently, however, they have been identified in tissues where they were not initially suspected to reside, which raises the following question: what external and internal factors dictate differentiation toward this specific role? In this discussion, we will focus on murine studies to determine how these cells are identified (e.g., markers and function) and ask the broader question of factors triggering M-cell localization and patterning. Then, through the consideration of unconventional M cells, which include villous M cells, Type II taste cells, and medullary thymic epithelial M cells (microfold mTECs), we will establish the M cell as not just a player in mucosal immunity but as a versatile niche cell that adapts to its home tissue. To this end, we will consider the lymphoid structure relationship and apical stimuli to better discuss how the differing cellular programming and the physical environment within each tissue yield these cells and their unique organization. Thus, by exploring this constellation of M cells, we hope to better understand the multifaceted nature of this cell in its different anatomical locales.


Sujet(s)
Immunité muqueuse , Animaux , Souris , Tissu lymphoïde/immunologie , Tissu lymphoïde/cytologie , Humains , Cellules épithéliales/immunologie , Différenciation cellulaire , Muqueuse intestinale/immunologie , Muqueuse intestinale/cytologie , Niche de cellules souches , Cellules M
11.
Front Cell Infect Microbiol ; 14: 1394070, 2024.
Article de Anglais | MEDLINE | ID: mdl-38895731

RÉSUMÉ

Mycobacterium avium subsp. paratuberculosis (Map) is the etiological agent of paratuberculosis (PTB), a chronic intestinal inflammatory disease that causes high economical losses in dairy livestock worldwide. Due to the absence of widely available preventive or therapeutical treatments, new alternative therapies are needed. In this study, the effect of a probiotic alone or in combination with a commercial vaccine has been evaluated in a rabbit model. Vaccination enhanced the humoral response, exerted a training effect of peripheral polymorphonuclear neutrophils (PMNs) against homologous and heterologous stimuli, stimulated the release of pro-inflammatory cytokines by gut-associated lymphoid tissue (GALT) macrophages, and reduced the bacterial burden in GALT as well. However, the administration of the probiotic after vaccination did not affect the PMN activity, increased metabolic demand, and supressed pro-inflammatory cytokines, although humoral response and bacterial burden decrease in GALT was maintained similar to vaccination alone. The administration of the probiotic alone did not enhance the humoral response or PMN activity, and the bacterial burden in GALT was further increased compared to the only challenged group. In conclusion, the probiotic was able to modulate the immune response hampering the clearance of the infection and was also able to affect the response of innate immune cells after vaccination. This study shows that the administration of a probiotic can modulate the immune response pathways triggered by vaccination and/or infection and even exacerbate the outcome of the disease, bringing forward the importance of verifying treatment combinations in the context of each particular infectious agent.


Sujet(s)
Cytokines , Mycobacterium avium ssp. paratuberculosis , Granulocytes neutrophiles , Paratuberculose , Probiotiques , Vaccination , Animaux , Probiotiques/administration et posologie , Paratuberculose/prévention et contrôle , Paratuberculose/immunologie , Paratuberculose/microbiologie , Mycobacterium avium ssp. paratuberculosis/immunologie , Lapins , Granulocytes neutrophiles/immunologie , Cytokines/métabolisme , Vaccins antibactériens/immunologie , Vaccins antibactériens/administration et posologie , Macrophages/immunologie , Modèles animaux de maladie humaine , Tissu lymphoïde/immunologie , Tissu lymphoïde/microbiologie , Femelle , Immunité humorale , Anticorps antibactériens/sang
12.
Infect Immun ; 92(7): e0026323, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38899881

RÉSUMÉ

Because most humans resist Mycobacterium tuberculosis infection, there is a paucity of lung samples to study. To address this gap, we infected Diversity Outbred mice with M. tuberculosis and studied the lungs of mice in different disease states. After a low-dose aerosol infection, progressors succumbed to acute, inflammatory lung disease within 60 days, while controllers maintained asymptomatic infection for at least 60 days, and then developed chronic pulmonary tuberculosis (TB) lasting months to more than 1 year. Here, we identified features of asymptomatic M. tuberculosis infection by applying computational and statistical approaches to multimodal data sets. Cytokines and anti-M. tuberculosis cell wall antibodies discriminated progressors vs controllers with chronic pulmonary TB but could not classify mice with asymptomatic infection. However, a novel deep-learning neural network trained on lung granuloma images was able to accurately classify asymptomatically infected lungs vs acute pulmonary TB in progressors vs chronic pulmonary TB in controllers, and discrimination was based on perivascular and peribronchiolar lymphocytes. Because the discriminatory lesion was rich in lymphocytes and CD4 T cell-mediated immunity is required for resistance, we expected CD4 T-cell genes would be elevated in asymptomatic infection. However, the significantly different, highly expressed genes were from B-cell pathways (e.g., Bank1, Cd19, Cd79, Fcmr, Ms4a1, Pax5, and H2-Ob), and CD20+ B cells were enriched in the perivascular and peribronchiolar regions of mice with asymptomatic M. tuberculosis infection. Together, these results indicate that genetically controlled B-cell responses are important for establishing asymptomatic M. tuberculosis lung infection.


Sujet(s)
Lymphocytes B , Poumon , Mycobacterium tuberculosis , Tuberculose pulmonaire , Animaux , Souris , Tuberculose pulmonaire/immunologie , Tuberculose pulmonaire/microbiologie , Tuberculose pulmonaire/anatomopathologie , Mycobacterium tuberculosis/immunologie , Lymphocytes B/immunologie , Poumon/microbiologie , Poumon/anatomopathologie , Poumon/immunologie , Granulome/microbiologie , Granulome/immunologie , Granulome/anatomopathologie , Tissu lymphoïde/immunologie , Tissu lymphoïde/microbiologie , Tissu lymphoïde/anatomopathologie , Modèles animaux de maladie humaine , Femelle , Infections asymptomatiques , Cytokines/métabolisme , Cytokines/génétique
13.
Sci Adv ; 10(22): eadn7786, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38809992

RÉSUMÉ

Viruses, bacteria, and parasites frequently cause infections in the gastrointestinal tract, but traditional vaccination strategies typically elicit little or no mucosal antibody responses. Here, we report a strategy to effectively concentrate immunogens and adjuvants in gut-draining lymph nodes (LNs) to induce gut-associated mucosal immunity. We prepared nanoemulsions (NEs) based on biodegradable oils commonly used as vaccine adjuvants, which encapsulated a potent Toll-like receptor agonist and displayed antigen conjugated to their surface. Following intraperitoneal administration, these NEs accumulated in gut-draining mesenteric LNs, priming strong germinal center responses and promoting B cell class switching to immunoglobulin A (IgA). Optimized NEs elicited 10- to 1000-fold higher antigen-specific IgG and IgA titers in the serum and feces, respectively, compared to free antigen mixed with NE, and strong neutralizing antibody titers against severe acute respiratory syndrome coronavirus 2. Thus, robust gut humoral immunity can be elicited by exploiting the unique lymphatic collection pathways of the gut with a lymph-targeting vaccine formulation.


Sujet(s)
Immunité humorale , Animaux , Souris , Tube digestif/immunologie , Tissu lymphoïde/immunologie , Immunité muqueuse/effets des médicaments et des substances chimiques , SARS-CoV-2/immunologie , COVID-19/prévention et contrôle , COVID-19/immunologie , Anticorps antiviraux/immunologie , Noeuds lymphatiques/immunologie , Immunoglobuline A/immunologie , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/administration et posologie , Anticorps neutralisants/immunologie , Femelle , Lymphocytes B/immunologie , Adjuvants vaccinaux , Souris de lignée C57BL , Humains
14.
Front Immunol ; 15: 1377913, 2024.
Article de Anglais | MEDLINE | ID: mdl-38799420

RÉSUMÉ

Introduction: The atypical chemokine receptor 2 (ACKR2) is a chemokine scavenger receptor, which limits inflammation and organ damage in several experimental disease models including kidney diseases. However, potential roles of ACKR2 in reducing inflammation and tissue injury in autoimmune disorders like systemic lupus erythematosus (SLE) and lupus nephritis are unknown, as well as its effects on systemic autoimmunity. Methods: To characterize functional roles of ACKR2 in SLE, genetic Ackr2 deficiency was introduced into lupus-prone C57BL/6lpr (Ackr2-/- B6lpr) mice. Results: Upon inflammatory stimulation in vitro, secreted chemokine levels increased in Ackr2 deficient tubulointerstitial tissue but not glomeruli. Moreover, Ackr2 expression was induced in kidneys and lungs of female C57BL/6lpr mice developing SLE. However, female Ackr2-/- B6lpr mice at 28 weeks of age showed similar renal functional parameters as wildtype (WT)-B6lpr mice. Consistently, assessment of activity and chronicity indices for lupus nephritis revealed comparable renal injury. Interestingly, Ackr2-/- B6lpr mice showed significantly increased renal infiltrates of CD3+ T and B cells, but not neutrophils, macrophages or dendritic cells, with T cells predominantly accumulating in the tubulointerstitial compartment of Ackr2-/- B6lpr mice. In addition, histology demonstrated significantly increased peribronchial lung infiltrates of CD3+ T cells in Ackr2-/- B6lpr mice. Despite this, protein levels of pro-inflammatory chemokines and mRNA expression of inflammatory mediators were not different in kidneys and lungs of WT- and Ackr2-/- B6lpr mice. This data suggests compensatory mechanisms for sufficient chemokine clearance in Ackr2-deficient B6lpr mice in vivo. Analysis of systemic autoimmune responses revealed comparable levels of circulating lupus-associated autoantibodies and glomerular immunoglobulin deposition in the two genotypes. Interestingly, similar to kidney and lung CD4+ T cell numbers and activation were significantly increased in spleens of Ackr2-deficient B6lpr mice. In lymph nodes of Ackr2-/- B6lpr mice abundance of activated dendritic cells decreased, but CD4+ T cell numbers were comparable to WT. Moreover, increased plasma levels of CCL2 were present in Ackr2-/- B6lpr mice, which may facilitate T cell mobilization into spleens and peripheral organs. Discussion: In summary, we show that ACKR2 prevents expansion of T cells and formation of tertiary lymphoid tissue, but is not essential to limit autoimmune tissue injury in lupus-prone B6lpr mice.


Sujet(s)
Lupus érythémateux disséminé , Glomérulonéphrite lupique , Souris de lignée C57BL , Souris knockout , Lymphocytes T , Structures lymphoïdes tertiaires , Animaux , Souris , Femelle , Lupus érythémateux disséminé/immunologie , Structures lymphoïdes tertiaires/immunologie , Glomérulonéphrite lupique/immunologie , Glomérulonéphrite lupique/métabolisme , Glomérulonéphrite lupique/anatomopathologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Modèles animaux de maladie humaine , Rein/anatomopathologie , Rein/immunologie , Rein/métabolisme , Auto-immunité , Système Duffy/génétique , Tissu lymphoïde/immunologie , Tissu lymphoïde/métabolisme , Prolifération cellulaire ,
15.
Immunol Rev ; 324(1): 78-94, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38717136

RÉSUMÉ

It is now widely understood that visceral adipose tissue (VAT) is a highly active and dynamic organ, with many functions beyond lipid accumulation and storage. In this review, we discuss the immunological role of this tissue, underpinned by the presence of fat-associated lymphoid clusters (FALCs). FALC's distinctive structure and stromal cell composition support a very different immune cell mix to that found in classical secondary lymphoid organs, which underlies their unique functions of filtration, surveillance, innate-like immune responses, and adaptive immunity within the serous cavities. FALCs are important B cell hubs providing B1 cell-mediated frontline protection against infection and supporting B2 cell-adaptative immune responses. Beyond these beneficial immune responses orchestrated by FALCs, immune cells within VAT play important homeostatic role. Dysregulation of immune cells during obesity and aging leads to chronic pathological "metabolic inflammation", which contributes to the development of cardiometabolic diseases. Here, we examine the emerging and complex functions of B cells in VAT homeostasis and the metabolic complications of obesity, highlighting the potential role that FALCs play and emphasize the areas where further research is needed.


Sujet(s)
Lymphocytes B , Homéostasie , Graisse intra-abdominale , Humains , Animaux , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Graisse intra-abdominale/immunologie , Graisse intra-abdominale/métabolisme , Obésité/immunologie , Obésité/métabolisme , Tissu lymphoïde/immunologie , Tissu lymphoïde/métabolisme , Immunité acquise
16.
Nat Commun ; 15(1): 4051, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38744839

RÉSUMÉ

Intestinal homeostasis is maintained by the response of gut-associated lymphoid tissue to bacteria transported across the follicle associated epithelium into the subepithelial dome. The initial response to antigens and how bacteria are handled is incompletely understood. By iterative application of spatial transcriptomics and multiplexed single-cell technologies, we identify that the double negative 2 subset of B cells, previously associated with autoimmune diseases, is present in the subepithelial dome in health. We show that in this location double negative 2 B cells interact with dendritic cells co-expressing the lupus autoantigens DNASE1L3 and C1q and microbicides. We observe that in humans, but not in mice, dendritic cells expressing DNASE1L3 are associated with sampled bacteria but not DNA derived from apoptotic cells. We propose that fundamental features of autoimmune diseases are microbiota-associated, interacting components of normal intestinal immunity.


Sujet(s)
Lymphocytes B , Cellules dendritiques , Endodeoxyribonucleases , Microbiome gastro-intestinal , Animaux , Femelle , Humains , Mâle , Souris , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Endodeoxyribonucleases/métabolisme , Endodeoxyribonucleases/génétique , Microbiome gastro-intestinal/immunologie , Muqueuse intestinale/immunologie , Muqueuse intestinale/microbiologie , Muqueuse intestinale/métabolisme , Tissu lymphoïde/immunologie , Tissu lymphoïde/métabolisme , Souris de lignée C57BL
17.
Mod Pathol ; 37(8): 100512, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38734236

RÉSUMÉ

This review focuses on mature T cells, natural killer (NK) cells, and stroma-derived neoplasms in the fifth edition of the World Health Organization classification of hematolymphoid tumors, including changes from the revised fourth edition. Overall, information has expanded, primarily due to advancements in genomic understanding. The updated classification adopts a hierarchical format. The updated classification relies on a multidisciplinary approach, incorporating insights from a diverse group of pathologists, clinicians, and geneticists. Indolent NK-cell lymphoproliferative disorder of the gastrointestinal tract, Epstein-Barr virus-positive nodal T- and NK-cell lymphoma, and several stroma-derived neoplasms of lymphoid tissues have been newly introduced or included. The review also provides guidance on how the fifth edition of the World Health Organization classification of hematolymphoid tumors can be applied in routine clinical practice.


Sujet(s)
Cellules tueuses naturelles , Organisation mondiale de la santé , Humains , Cellules tueuses naturelles/anatomopathologie , Cellules tueuses naturelles/immunologie , Lymphocytes T/immunologie , Lymphocytes T/anatomopathologie , Tissu lymphoïde/anatomopathologie , Tissu lymphoïde/immunologie , Cellules stromales/anatomopathologie , Cellules stromales/immunologie , Tumeurs hématologiques/anatomopathologie , Tumeurs hématologiques/classification , Tumeurs hématologiques/immunologie
18.
Retrovirology ; 21(1): 8, 2024 May 02.
Article de Anglais | MEDLINE | ID: mdl-38693565

RÉSUMÉ

The study of HIV infection and pathogenicity in physical reservoirs requires a biologically relevant model. The human immune system (HIS) mouse is an established model of HIV infection, but defects in immune tissue reconstitution remain a challenge for examining pathology in tissues. We utilized exogenous injection of the human recombinant FMS-like tyrosine kinase 3 ligand (rFLT-3 L) into the hematopoietic stem cell (HSC) cord blood HIS mouse model to significantly expand the total area of lymph node (LN) and the number of circulating human T cells. The results enabled visualization and quantification of HIV infectivity, CD4 T cell depletion and other measures of pathogenesis in the secondary lymphoid tissues of the spleen and LN. Treatment with the Caspase-1/4 inhibitor VX-765 limited CD4+ T cell loss in the spleen and reduced viral load in both the spleen and axillary LN. In situ hybridization further demonstrated a decrease in viral RNA in both the spleen and LN. Transcriptomic analysis revealed that in vivo inhibition of caspase-1/4 led to an upregulation in host HIV restriction factors including SAMHD1 and APOBEC3A. These findings highlight the use of rFLT-3 L to augment human immune system characteristics in HIS mice to support investigations of HIV pathogenesis and test host directed therapies, though further refinements are needed to further augment LN architecture and cellular populations. The results further provide in vivo evidence of the potential to target inflammasome pathways as an avenue of host-directed therapy to limit immune dysfunction and virus replication in tissue compartments of HIV+ persons.


Sujet(s)
Lymphocytes T CD4+ , Modèles animaux de maladie humaine , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Animaux , Souris , Infections à VIH/immunologie , Infections à VIH/virologie , Infections à VIH/traitement médicamenteux , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Humains , Lymphocytes T CD4+/immunologie , Tissu lymphoïde/virologie , Tissu lymphoïde/immunologie , Charge virale/effets des médicaments et des substances chimiques , Rate/virologie , Rate/immunologie , Noeuds lymphatiques/immunologie , Noeuds lymphatiques/virologie , Caspases/métabolisme , Inhibiteurs des caspases/pharmacologie , Antirétroviraux/usage thérapeutique
19.
Fish Shellfish Immunol ; 149: 109535, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38582231

RÉSUMÉ

Mucosal immunity in mucosa-associated lymphoid tissues (MALTs) plays crucial roles in resisting infection by pathogens, including parasites, bacteria and viruses. However, the mucosal immune response in the MALTs of large yellow croaker (Larimichthys crocea) upon parasitic infection remains largely unknown. In this study, we investigated the role of B cells and T cells in the MALTs of large yellow croaker following Cryptocaryon irritans infection. Upon C. irritans infection, the total IgM and IgT antibody levels were significantly increased in the skin mucus and gill mucus. Notably, parasite-specific IgM antibody level was increased in the serum, skin and gill mucus following parasitic infection, while the level of parasite-specific IgT antibody was exclusively increased in MALTs. Moreover, parasitic infection induced both local and systemic aggregation and proliferation of IgM+ B cells, suggesting that the increased levels of IgM in mucus may be derived from both systemic and mucosal immune tissues. In addition, we observed significant aggregation and proliferation of T cells in the gill, head kidney and spleen, suggesting that T cells may also be involved in the systemic and mucosal immune responses upon parasitic infection. Overall, our findings provided further insights into the role of immunoglobulins against pathogenic infection, and the simultaneous aggregation and proliferation of both B cells and T cells at mucosal surfaces suggested potential interactions between these two major lymphocyte populations during parasitic infection.


Sujet(s)
Lymphocytes B , Infections à ciliophores , Ciliophora , Maladies des poissons , Perciformes , Lymphocytes T , Animaux , Maladies des poissons/immunologie , Maladies des poissons/parasitologie , Perciformes/immunologie , Infections à ciliophores/médecine vétérinaire , Infections à ciliophores/immunologie , Lymphocytes B/immunologie , Ciliophora/physiologie , Lymphocytes T/immunologie , Immunité muqueuse , Tissu lymphoïde/immunologie , Immunoglobuline M/immunologie , Immunoglobuline M/sang , Prolifération cellulaire
20.
Immunol Rev ; 324(1): 68-77, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38662554

RÉSUMÉ

The milky spots in omentum are atypical lymphoid tissues that play a pivotal role in regulating immune responses in the peritoneal cavity. The milky spots act as central hubs for collecting antigens and particles from the peritoneal cavity, regulating lymphocyte trafficking, promoting the differentiation and self-renewal of immune cells, and supporting the local germinal centre response. In addition, the milky spots exhibit unique developmental characteristics that combine the features of secondary and tertiary lymphoid tissues. These structures are innately programmed to form during foetal development; however, they can also be formed postnatally in response to peritoneal irritation such as inflammation, infection, obesity, or tumour metastasis. In this review, I discuss emerging perspectives on homeostatic development and organization of the milky spots.


Sujet(s)
Omentum , Humains , Animaux , Omentum/immunologie , Différenciation cellulaire , Homéostasie , Tissu lymphoïde/immunologie , Centre germinatif/immunologie , Cavité péritonéale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE