Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 13.289
Filtrar
1.
Front Immunol ; 15: 1419527, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39286244

RESUMO

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.


Assuntos
Imunidade nas Mucosas , Imunomodulação , Humanos , Animais , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Tecido Linfoide/imunologia , Vacinas/imunologia , Mucosa Nasal/imunologia , Mucosa Nasal/metabolismo , Administração Intranasal
2.
Int Rev Cell Mol Biol ; 388: 1-19, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39260934

RESUMO

Chemokine receptors are a complex superfamily of surface G protein-coupled receptors present mostly in leukocytes. In this chapter, we review the presence and functions of chemokine receptors in the immune cells of the primary and secondary lymphoid organs. Those include bone marrow, thymus, spleen, lymph nodes, and Peyer's patches as the main components of the gut-associated lymphoid tissue. There are general groups of chemokine receptors: conventional and atypical. We will mostly cover the role of conventional chemokine receptors, which are divided into four classes (CC, CXC, CX3C, and XC). Some relevant members are CXCR4, CXCR5, CCR4 and CCR7. For example, CXCR4 is a key chemokine receptor in the bone marrow controlling from the homing of progenitor cells into the bone marrow, the development of B cells, to the homing of long-lived plasma cells to this primary lymphoid organ. CCR7 and CCR4 are two of the main players in the thymus. CCR7 along with CCR9 control the traffic of thymic seed progenitors into the thymus, while CCR4 and CCR7 are critical for the entry of thymocytes into the medulla and as controllers of the central tolerance in the thymus. CXCR4 and CXCR5 have major roles in the spleen, guiding the maturation and class-switching of B cells in the different areas of the germinal center. In the T-cell zone, CCR7 guides the differentiation of naïve T cells. CCR7 also controls and directs the entry of T cells, B cells, and dendritic cells into secondary lymphoid tissues, including the spleen and lymph nodes. As new technologies emerge, techniques such as high dimensional spectral flow cytometry or single-cell sequencing allow a more comprehensive knowledge of the chemokine receptor network and their ligands, as well as the discovery of new interactions mediating unknown and overlooked mechanisms in health and disease.


Assuntos
Tecido Linfoide , Receptores de Quimiocinas , Humanos , Animais , Receptores de Quimiocinas/metabolismo , Tecido Linfoide/metabolismo , Timo/metabolismo , Timo/citologia
3.
J Exp Med ; 221(10)2024 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-39240335

RESUMO

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.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Células B de Memória , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Vacinas de mRNA , Humanos , Vacinas contra COVID-19/imunologia , Vacinas de mRNA/imunologia , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/genética , Células B de Memória/imunologia , COVID-19/prevenção & controle , COVID-19/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Tecido Linfoide/imunologia , Dispositivos Lab-On-A-Chip , Vacinas Sintéticas/imunologia , RNA Mensageiro/genética , RNA Mensageiro/imunologia , RNA Mensageiro/metabolismo , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Lipossomos , Nanopartículas
4.
J Exp Med ; 221(9)2024 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-39093311

RESUMO

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.


Assuntos
Movimento Celular , Tecido Linfoide , Humanos , Tecido Linfoide/imunologia , Tecido Linfoide/citologia , Movimento Celular/imunologia , Linfócitos B/imunologia , Imunoglobulina M/metabolismo , Imunoglobulina M/imunologia , Subpopulações de Linfócitos B/imunologia , Células Precursoras de Linfócitos B/imunologia , Células Precursoras de Linfócitos B/citologia , Diferenciação Celular/imunologia
5.
Signal Transduct Target Ther ; 9(1): 225, 2024 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-39198425

RESUMO

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.


Assuntos
Doenças Autoimunes , Estruturas Linfoides Terciárias , Humanos , Estruturas Linfoides Terciárias/imunologia , Estruturas Linfoides Terciárias/patologia , Estruturas Linfoides Terciárias/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/genética , Doenças Autoimunes/terapia , Doenças Autoimunes/patologia , Neoplasias/imunologia , Neoplasias/terapia , Neoplasias/genética , Neoplasias/patologia , Inflamação/imunologia , Inflamação/genética , Inflamação/patologia , Tecido Linfoide/imunologia , Tecido Linfoide/patologia , Animais , Citocinas/imunologia , Citocinas/genética
6.
Chem Soc Rev ; 53(15): 7657-7680, 2024 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-38958009

RESUMO

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.


Assuntos
Imunomodulação , Inflamação , Nanoestruturas , Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Nanoestruturas/química , Inflamação/tratamento farmacológico , Inflamação/imunologia , Imunomodulação/efeitos dos fármacos , Animais , Imunoterapia , Tecido Linfoide/imunologia , Tecido Linfoide/efeitos dos fármacos
7.
Nature ; 632(8025): 637-646, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39085603

RESUMO

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.


Assuntos
Imunoglobulina A , Tecido Linfoide , Mucosa Nasal , Plasmócitos , Linfócitos T , Conchas Nasais , Animais , Feminino , Masculino , Camundongos , Bactérias/imunologia , Movimento Celular , Quimiocinas CC/imunologia , Quimiocinas CC/metabolismo , Centro Germinativo/imunologia , Centro Germinativo/citologia , Imunoglobulina A/imunologia , Imunoglobulina A/metabolismo , Tecido Linfoide/imunologia , Tecido Linfoide/citologia , Camundongos Endogâmicos C57BL , Mucosa Nasal/citologia , Mucosa Nasal/imunologia , Plasmócitos/imunologia , Plasmócitos/citologia , Plasmócitos/metabolismo , Linfócitos T/imunologia , Linfócitos T/citologia , Linfócitos T/metabolismo , Conchas Nasais/citologia , Conchas Nasais/imunologia , Vacinação , Administração Intranasal , Vacinas/imunologia , Simbiose
8.
Sci Rep ; 14(1): 15535, 2024 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-38969759

RESUMO

Since PCV4 was first described in 2019, the virus has been identified in several countries in Southeast Asia and Europe. Most studies have been limited to detecting PCV4 by PCR. Thus, PCV4 has an unclear association with clinical disease. This study utilized 512 porcine clinical lung, feces, spleen, serum, lymphoid tissue, and fetus samples submitted to the ISU-VDL from June-September 2023. PCV4 was detected in 8.6% of samples with an average Ct value of 33. While detection rates among sample types were variable, lymphoid tissue had the highest detection rate (18.7%). Two ORF2 sequences were obtained from lymphoid tissue samples and had 96.36-98.98% nucleotide identity with reference sequences. Direct detection of PCV4 by RNAscope revealed viral replication in B lymphocytes and macrophages in lymph node germinal centers and histiocytic and T lymphocyte infiltration in the lamina propria of the small intestine. PCV4 detection was most commonly observed in nursery to finishing aged pigs displaying respiratory and enteric disease. Coinfection with PCV2, PCV3, and other endemic pathogens was frequently observed, highlighting the complex interplay between different PCVs and their potential roles in disease pathogenesis. This study provides insights into the frequency of detection, tissue distribution, and genetic characteristics of PCV4 in the US.


Assuntos
Infecções por Circoviridae , Circovirus , Doenças dos Suínos , Animais , Circovirus/genética , Circovirus/isolamento & purificação , Suínos , Infecções por Circoviridae/veterinária , Infecções por Circoviridae/virologia , Doenças dos Suínos/virologia , Estados Unidos/epidemiologia , Tecido Linfoide/virologia , Coinfecção/virologia , Coinfecção/veterinária , Pulmão/virologia
9.
PLoS One ; 19(7): e0292408, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38950025

RESUMO

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.


Assuntos
Coinfecção , Citocinas , Nematospiroides dubius , Toxoplasma , Animais , Coinfecção/imunologia , Coinfecção/parasitologia , Toxoplasma/imunologia , Camundongos , Citocinas/metabolismo , Nematospiroides dubius/imunologia , Infecções por Strongylida/imunologia , Infecções por Strongylida/parasitologia , Infecções por Strongylida/mortalidade , Toxoplasmose/imunologia , Toxoplasmose/mortalidade , Toxoplasmose/complicações , Feminino , Toxoplasmose Animal/imunologia , Toxoplasmose Animal/mortalidade , Toxoplasmose Animal/parasitologia , Baço/imunologia , Baço/patologia , Baço/parasitologia , Carga Parasitária , Tecido Linfoide/imunologia , Tecido Linfoide/patologia , Tecido Linfoide/parasitologia
10.
Vet Immunol Immunopathol ; 274: 110785, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38861830

RESUMO

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.


Assuntos
Vacinas contra Influenza , Infecções por Orthomyxoviridae , Doenças dos Suínos , Animais , Suínos/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/veterinária , Infecções por Orthomyxoviridae/prevenção & controle , Doenças dos Suínos/imunologia , Doenças dos Suínos/virologia , Doenças dos Suínos/prevenção & controle , Linfócitos T Auxiliares-Indutores/imunologia , Sistema Respiratório/imunologia , Sistema Respiratório/virologia , Tecido Linfoide/imunologia , Memória Imunológica , Células T de Memória/imunologia , Subpopulações de Linfócitos T/imunologia , Vírus da Influenza A/imunologia , Vacinação/veterinária
11.
Infect Immun ; 92(7): e0026323, 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38899881

RESUMO

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.


Assuntos
Linfócitos B , Pulmão , Mycobacterium tuberculosis , Tuberculose Pulmonar , Animais , Camundongos , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/microbiologia , Tuberculose Pulmonar/patologia , Mycobacterium tuberculosis/imunologia , Linfócitos B/imunologia , Pulmão/microbiologia , Pulmão/patologia , Pulmão/imunologia , Granuloma/microbiologia , Granuloma/imunologia , Granuloma/patologia , Tecido Linfoide/imunologia , Tecido Linfoide/microbiologia , Tecido Linfoide/patologia , Modelos Animais de Doenças , Feminino , Infecções Assintomáticas , Citocinas/metabolismo , Citocinas/genética
12.
Front Cell Infect Microbiol ; 14: 1394070, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38895731

RESUMO

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.


Assuntos
Citocinas , Mycobacterium avium subsp. paratuberculosis , Neutrófilos , Paratuberculose , Probióticos , Vacinação , Animais , Probióticos/administração & dosagem , Paratuberculose/prevenção & controle , Paratuberculose/imunologia , Paratuberculose/microbiologia , Mycobacterium avium subsp. paratuberculosis/imunologia , Coelhos , Neutrófilos/imunologia , Citocinas/metabolismo , Vacinas Bacterianas/imunologia , Vacinas Bacterianas/administração & dosagem , Macrófagos/imunologia , Modelos Animais de Doenças , Tecido Linfoide/imunologia , Tecido Linfoide/microbiologia , Feminino , Imunidade Humoral , Anticorpos Antibacterianos/sangue
13.
Ultrasound Med Biol ; 50(8): 1247-1254, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38834492

RESUMO

OBJECTIVE: Needle biopsy is a common technique used to obtain cell and tissue samples for diagnostics. Currently, two biopsy methods are widely used: (i) fine-needle aspiration biopsy (FNAB) and (ii) core needle biopsy (CNB). However, these methods have limitations. Recently, we developed ultrasound-enhanced fine-needle aspiration biopsy (USeFNAB), which employs a needle that flexurally oscillates at an ultrasonic frequency of ∼32 kHz. The needle motion contributes to increased tissue collection while preserving cells and tissue constructs for pathological assessment. Previously, USeFNAB has been investigated only in ex vivo animal tissue. The present study was aimed at determining the feasibility of using USeFNAB in human epithelial and lymphoid tissue. METHODS: Needle biopsy samples were acquired using FNAB, CNB and USeFNAB on ex vivo human tonsils (N = 10). The tissue yield and quality were quantified by weight measurement and blinded pathologists' assessments. The biopsy methods were then compared. RESULTS: The results revealed sample mass increases of, on average, 2.3- and 5.4-fold with USeFNAB compared with the state-of-the-art FNAB and CNB, respectively. The quality of tissue fragments collected by USeFNAB was equivalent to that collected by the state-of-the-art methods in terms of morphology and immunohistochemical stainings made from cell blocks as judged by pathologists. CONCLUSION: Our study indicates that USeFNAB is a promising method that could improve tissue yield to ensure sufficient material for ancillary histochemical and molecular studies for diagnostic pathology, thereby potentially increasing diagnostic accuracy.


Assuntos
Tecido Linfoide , Tonsila Palatina , Humanos , Tonsila Palatina/patologia , Tonsila Palatina/diagnóstico por imagem , Tecido Linfoide/patologia , Tecido Linfoide/diagnóstico por imagem , Biópsia por Agulha Fina/métodos , Estudos de Viabilidade , Ultrassonografia de Intervenção/métodos , Biópsia Guiada por Imagem/métodos , Epitélio/patologia
14.
Front Immunol ; 15: 1400739, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38863701

RESUMO

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.


Assuntos
Imunidade nas Mucosas , Animais , Camundongos , Tecido Linfoide/imunologia , Tecido Linfoide/citologia , Humanos , Células Epiteliais/imunologia , Diferenciação Celular , Mucosa Intestinal/imunologia , Mucosa Intestinal/citologia , Nicho de Células-Tronco , Células M
15.
Mod Pathol ; 37(8): 100512, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38734236

RESUMO

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.


Assuntos
Células Matadoras Naturais , Organização Mundial da Saúde , Humanos , Células Matadoras Naturais/patologia , Células Matadoras Naturais/imunologia , Linfócitos T/imunologia , Linfócitos T/patologia , Tecido Linfoide/patologia , Tecido Linfoide/imunologia , Células Estromais/patologia , Células Estromais/imunologia , Neoplasias Hematológicas/patologia , Neoplasias Hematológicas/classificação , Neoplasias Hematológicas/imunologia
16.
Trop Anim Health Prod ; 56(5): 179, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38809309

RESUMO

We evaluated the effects of supplementing yeast mannan-reach-fraction on growth performance, jejunal morphology and lymphoid tissue characteristics in weaned piglets challenged with E. Coli F4. A total of 20 crossbred piglets were used. At weaning, piglets were assigned at random to one of four groups: piglets challenged and fed the basal diet supplemented with yeast mannan-rich fraction (C-MRF, n = 5); piglets challenged and fed the basal diet (C-BD, n = 5); piglets not challenged and fed the basal diet supplemented with yeast mannan-rich fraction (NC-MRF, n = 5), and piglets not challenged and fed the basal diet (NC-BD). Each dietary treatment had five replicates. On days 4, 5 and 10, piglets were orally challenged with 108 CFU/mL of E. Coli F4. C-MRF piglets had higher BW (p = 0.002; interactive effect) than C-BD piglets. C-MRF piglets had higher (p = 0.02; interactive effect) ADG in comparison with C-BD piglets. C-MRF piglets had higher (p = 0.04; interactive effect) ADFI than C-BD piglets. The diameter of lymphoid follicles was larger (p = 0.010; interactive effect) in the tonsils of C-MRF piglets than C-BD piglets. Lymphoid cells proliferation was greater in the mesenteric lymphnodes and ileum (p = 0.04 and p = 0.03, respectively) of C-MRF piglets. A reduction (p > 0.05) in E. Coli adherence in the ileum of piglets fed MRF was observed. In conclusion, the results of the present study demonstrate that dietary yeast mannan-rich fraction supplementation was effective in protecting weaned piglets against E. Coli F4 challenge.


Assuntos
Suplementos Nutricionais , Escherichia coli Enterotoxigênica , Mananas , Leveduras , Animais , Suínos/crescimento & desenvolvimento , Suínos/microbiologia , Infecções por Escherichia coli/veterinária , Doenças dos Suínos/microbiologia , Jejuno/crescimento & desenvolvimento , Desmame , Criação de Animais Domésticos , Tecido Linfoide/fisiologia
17.
Immunol Rev ; 324(1): 78-94, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38717136

RESUMO

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.


Assuntos
Linfócitos B , Homeostase , Gordura Intra-Abdominal , Humanos , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Gordura Intra-Abdominal/imunologia , Gordura Intra-Abdominal/metabolismo , Obesidade/imunologia , Obesidade/metabolismo , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Imunidade Adaptativa
19.
Nat Commun ; 15(1): 4051, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38744839

RESUMO

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.


Assuntos
Linfócitos B , Células Dendríticas , Endodesoxirribonucleases , Microbioma Gastrointestinal , Animais , Feminino , Humanos , Masculino , Camundongos , Linfócitos B/imunologia , Linfócitos B/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Endodesoxirribonucleases/metabolismo , Endodesoxirribonucleases/genética , Microbioma Gastrointestinal/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/metabolismo , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Camundongos Endogâmicos C57BL
20.
Sci Adv ; 10(22): eadn7786, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38809992

RESUMO

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.


Assuntos
Imunidade Humoral , Animais , Camundongos , Trato Gastrointestinal/imunologia , Tecido Linfoide/imunologia , Imunidade nas Mucosas/efeitos dos fármacos , SARS-CoV-2/imunologia , COVID-19/prevenção & controle , COVID-19/imunologia , Anticorpos Antivirais/imunologia , Linfonodos/imunologia , Imunoglobulina A/imunologia , Vacinas contra COVID-19/imunologia , Vacinas contra COVID-19/administração & dosagem , Anticorpos Neutralizantes/imunologia , Feminino , Linfócitos B/imunologia , Adjuvantes de Vacinas , Camundongos Endogâmicos C57BL , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA