Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38.774
Filtrar
1.
Cell Rep ; 43(4): 114088, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38602878

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) features an immunosuppressive tumor microenvironment (TME) that resists immunotherapy. Tumor-associated macrophages, abundant in the TME, modulate T cell responses. Bone marrow stromal antigen 2-positive (BST2+) macrophages increase in KrasG12D/+; Trp53R172H/+; Pdx1-Cre mouse models during PDAC progression. However, their role in PDAC remains elusive. Our findings reveal a negative correlation between BST2+ macrophage levels and PDAC patient prognosis. Moreover, an increased ratio of exhausted CD8+ T cells is observed in tumors with up-regulated BST2+ macrophages. Mechanistically, BST2+ macrophages secrete CXCL7 through the ERK pathway and bind with CXCR2 to activate the AKT/mTOR pathway, promoting CD8+ T cell exhaustion. The combined blockade of CXCL7 and programmed death-ligand 1 successfully decelerates tumor growth. Additionally, cGAS-STING pathway activation in macrophages induces interferon (IFN)α synthesis leading to BST2 overexpression in the PDAC TME. This study provides insights into IFNα-induced BST2+ macrophages driving an immune-suppressive TME through ERK-CXCL7 signaling to regulate CD8+ T cell exhaustion in PDAC.


Assuntos
Antígeno 2 do Estroma da Médula Óssea , Proteínas Ligadas por GPI , Interferon-alfa , Neoplasias Pancreáticas , Macrófagos Associados a Tumor , Animais , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/metabolismo , Humanos , Camundongos , Proteínas Ligadas por GPI/metabolismo , Macrófagos Associados a Tumor/metabolismo , Macrófagos Associados a Tumor/imunologia , Macrófagos Associados a Tumor/patologia , Interferon-alfa/metabolismo , Microambiente Tumoral/imunologia , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/imunologia , Carcinoma Ductal Pancreático/metabolismo , Transdução de Sinais , Antígenos CD/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Camundongos Endogâmicos C57BL , Linhagem Celular Tumoral , Tolerância Imunológica , Feminino
2.
World J Gastroenterol ; 30(13): 1791-1800, 2024 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-38659486

RESUMO

Liver transplantation (LT) has become the most efficient treatment for pediatric and adult end-stage liver disease and the survival time after transplantation is becoming longer due to the development of surgical techniques and perioperative management. However, long-term side-effects of immunosuppressants, like infection, metabolic disorders and malignant tumor are gaining more attention. Immune tolerance is the status in which LT recipients no longer need to take any immunosuppressants, but the liver function and intrahepatic histology maintain normal. The approaches to achieve immune tolerance after transplantation include spontaneous, operational and induced tolerance. The first two means require no specific intervention but withdrawing immunosuppressant gradually during follow-up. No clinical factors or biomarkers so far could accurately predict who are suitable for immunosuppressant withdraw after transplantation. With the understanding to the underlying mechanisms of immune tolerance, many strategies have been developed to induce tolerance in LT recipients. Cellular strategy is one of the most promising methods for immune tolerance induction, including chimerism induced by hematopoietic stem cells and adoptive transfer of regulatory immune cells. The safety and efficacy of various cell products have been evaluated by prospective preclinical and clinical trials, while obstacles still exist before translating into clinical practice. Here, we will summarize the latest perspectives and concerns on the clinical application of cellular strategies in LT recipients.


Assuntos
Imunossupressores , Transplante de Fígado , Humanos , Transplante de Fígado/efeitos adversos , Transplante de Fígado/métodos , Imunossupressores/uso terapêutico , Imunossupressores/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/métodos , Tolerância Imunológica/imunologia , Doença Hepática Terminal/cirurgia , Doença Hepática Terminal/imunologia , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Tolerância ao Transplante/imunologia , Transferência Adotiva/métodos , Sobrevivência de Enxerto/imunologia , Sobrevivência de Enxerto/efeitos dos fármacos , Animais , Resultado do Tratamento , Linfócitos T Reguladores/imunologia , Fígado/imunologia , Fígado/patologia , Fígado/cirurgia
3.
Front Immunol ; 15: 1359914, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38646539

RESUMO

Worldwide, gastrointestinal (GI) cancer is recognized as one of the leading malignancies diagnosed in both genders, with mortality largely attributed to metastatic dissemination. It has been identified that in GI cancer, a variety of signaling pathways and key molecules are modified, leading to the emergence of an immunotolerance phenotype. Such modifications are pivotal in the malignancy's evasion of immune detection. Thus, a thorough analysis of the pathways and molecules contributing to GI cancer's immunotolerance is vital for advancing our comprehension and propelling the creation of efficacious pharmacological treatments. In response to this necessity, our review illuminates a selection of groundbreaking cellular signaling pathways associated with immunotolerance in GI cancer, including the Phosphoinositide 3-kinases/Akt, Janus kinase/Signal Transducer and Activator of Transcription 3, Nuclear Factor kappa-light-chain-enhancer of activated B cells, Transforming Growth Factor-beta/Smad, Notch, Programmed Death-1/Programmed Death-Ligand 1, and Wingless and INT-1/beta-catenin-Interleukin 10. Additionally, we examine an array of pertinent molecules like Indoleamine-pyrrole 2,3-dioxygenase, Human Leukocyte Antigen G/E, Glycoprotein A Repetitions Predominant, Clever-1, Interferon regulatory factor 8/Osteopontin, T-cell immunoglobulin and mucin-domain containing-3, Carcinoembryonic antigen-related cell adhesion molecule 1, Cell division control protein 42 homolog, and caspases-1 and -12.


Assuntos
Neoplasias Gastrointestinais , Transdução de Sinais , Humanos , Neoplasias Gastrointestinais/imunologia , Neoplasias Gastrointestinais/patologia , Neoplasias Gastrointestinais/metabolismo , Animais , Metástase Neoplásica , Tolerância Imunológica , Evasão Tumoral
4.
Immun Inflamm Dis ; 12(4): e1246, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38668747

RESUMO

BACKGROUND: Cow's milk protein allergy (CMPA) remains relatively understudied in Latin America. METHODS: In this observational study, we enrolled 64 patients with a median age of 3 months, of whom 60% were male. Patients included had a history of IgE-mediated reactions with IgE sensitization or non-IgE-mediated reactions or symptoms following exposure to cow's milk. They underwent skin prick test, ImmunoCAP, fecal calprotectin (FC), and fecal eosinophil-derived neurotoxin (EDN), in addition to double-blinded placebo-controlled oral food challenges (DBPCFC), with clinical evolution and tolerance acquisition observed over 1 year. RESULTS: Malnutrition was present in 78.1% of patients, and 87.5% had a family history of atopy, with 51.6% receiving exclusive breastfeeding. Gastrointestinal manifestations were prevalent in 90.6% of patients, followed by dermatological manifestations (10.9%), with only 2 experiencing anaphylaxis. IgE-mediated CMPA was observed in only six patients. In those with non-IgE-mediated CMPA, FC had a median of 284 mg/dL (IQR: 138.5-415.5), while EDN had a median of 508.5 mg/dL (IQR: 160.25-868). One year after diagnosis, median FC significantly decreased (p < 0.0001), and malnutrition prevalence reduced to 17.1%. Moreover, 81% of patients acquired tolerance following DBPCFC, with 52% utilizing nutritional replacement formulas at diagnosis. Notably, 94% of those extensively hydrolyzed casein-based formulas achieved tolerance (p = 0.08). CONCLUSION: Our findings provide a foundational framework for future investigations into CMPA diagnosis, tolerance acquisition, and the utilization of hypoallergenic formulas tailored to the unique characteristics of our region.


Assuntos
Tolerância Imunológica , Imunoglobulina E , Hipersensibilidade a Leite , Proteínas do Leite , Testes Cutâneos , Humanos , Hipersensibilidade a Leite/imunologia , Hipersensibilidade a Leite/epidemiologia , Hipersensibilidade a Leite/diagnóstico , Hipersensibilidade a Leite/sangue , Masculino , Lactente , Feminino , Peru/epidemiologia , Proteínas do Leite/imunologia , Imunoglobulina E/imunologia , Imunoglobulina E/sangue , Animais , Alérgenos/imunologia , Bovinos , Fezes , Complexo Antígeno L1 Leucocitário/análise
5.
Nature ; 628(8009): 854-862, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38570678

RESUMO

The intestinal immune system is highly adapted to maintaining tolerance to the commensal microbiota and self-antigens while defending against invading pathogens1,2. Recognizing how the diverse network of local cells establish homeostasis and maintains it in the complex immune environment of the gut is critical to understanding how tolerance can be re-established following dysfunction, such as in inflammatory disorders. Although cell and molecular interactions that control T regulatory (Treg) cell development and function have been identified3,4, less is known about the cellular neighbourhoods and spatial compartmentalization that shapes microorganism-reactive Treg cell function. Here we used in vivo live imaging, photo-activation-guided single-cell RNA sequencing5-7 and spatial transcriptomics to follow the natural history of T cells that are reactive towards Helicobacter hepaticus through space and time in the settings of tolerance and inflammation. Although antigen stimulation can occur anywhere in the tissue, the lamina propria-but not embedded lymphoid aggregates-is the key microniche that supports effector Treg (eTreg) cell function. eTreg cells are stable once their niche is established; however, unleashing inflammation breaks down compartmentalization, leading to dominance of CD103+SIRPα+ dendritic cells in the lamina propria. We identify and validate the putative tolerogenic interaction between CD206+ macrophages and eTreg cells in the lamina propria and identify receptor-ligand pairs that are likely to govern the interaction. Our results reveal a spatial mechanism of tolerance in the lamina propria and demonstrate how knowledge of local interactions may contribute to the next generation of tolerance-inducing therapies.


Assuntos
Helicobacter hepaticus , Tolerância Imunológica , Cadeias alfa de Integrinas , Análise de Célula Única , Linfócitos T Reguladores , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/citologia , Animais , Camundongos , Cadeias alfa de Integrinas/metabolismo , Helicobacter hepaticus/imunologia , Tolerância Imunológica/imunologia , Feminino , Masculino , Antígenos CD/metabolismo , Receptores Imunológicos/metabolismo , Receptores Imunológicos/imunologia , Inflamação/imunologia , Inflamação/patologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/citologia , Intestinos/imunologia , Intestinos/citologia , Camundongos Endogâmicos C57BL , RNA-Seq , Transcriptoma , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Células Dendríticas/imunologia
6.
Nature ; 628(8009): 844-853, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38570685

RESUMO

Mitochondria are critical modulators of antiviral tolerance through the release of mitochondrial RNA and DNA (mtDNA and mtRNA) fragments into the cytoplasm after infection, activating virus sensors and type-I interferon (IFN-I) response1-4. The relevance of these mechanisms for mitochondrial diseases remains understudied. Here we investigated mitochondrial recessive ataxia syndrome (MIRAS), which is caused by a common European founder mutation in DNA polymerase gamma (POLG1)5. Patients homozygous for the MIRAS variant p.W748S show exceptionally variable ages of onset and symptoms5, indicating that unknown modifying factors contribute to disease manifestation. We report that the mtDNA replicase POLG1 has a role in antiviral defence mechanisms to double-stranded DNA and positive-strand RNA virus infections (HSV-1, TBEV and SARS-CoV-2), and its p.W748S variant dampens innate immune responses. Our patient and knock-in mouse data show that p.W748S compromises mtDNA replisome stability, causing mtDNA depletion, aggravated by virus infection. Low mtDNA and mtRNA release into the cytoplasm and a slow IFN response in MIRAS offer viruses an early replicative advantage, leading to an augmented pro-inflammatory response, a subacute loss of GABAergic neurons and liver inflammation and necrosis. A population databank of around 300,000 Finnish individuals6 demonstrates enrichment of immunodeficient traits in carriers of the POLG1 p.W748S mutation. Our evidence suggests that POLG1 defects compromise antiviral tolerance, triggering epilepsy and liver disease. The finding has important implications for the mitochondrial disease spectrum, including epilepsy, ataxia and parkinsonism.


Assuntos
Alelos , DNA Polimerase gama , DNA Mitocondrial , Humanos , DNA Polimerase gama/genética , DNA Polimerase gama/metabolismo , Animais , Camundongos , DNA Mitocondrial/genética , Masculino , Feminino , Imunidade Inata/genética , SARS-CoV-2/imunologia , Herpesvirus Humano 1/imunologia , Tolerância Imunológica/genética , COVID-19/imunologia , COVID-19/virologia , COVID-19/genética , Técnicas de Introdução de Genes , Mitocôndrias/metabolismo , Mutação
7.
Cancer Immunol Res ; 12(4): 393-399, 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38562083

RESUMO

CD4+CD25hiFOXP3+ regulatory T cells (Treg) play major roles in the maintenance of immune tolerance, prevention of inflammation, and tissue homeostasis and repair. In contrast with these beneficial roles, Tregs are abundant in virtually all tumors and have been mechanistically linked to disease progression, metastases development, and therapy resistance. Tregs are thus recognized as a major target for cancer immunotherapy. Compared with other sites in the body, tumors harbor hyperactivated Treg subsets whose molecular characteristics are only beginning to be elucidated. Here, we describe current knowledge of intratumoral Tregs and discuss their potential cellular and tissue origin. Furthermore, we describe currently recognized molecular regulators that drive differentiation and maintenance of Tregs in cancer, with a special focus on those signals regulating their chronic immune activation, with relevant implications for cancer progression and therapy.


Assuntos
Neoplasias , Linfócitos T Reguladores , Humanos , Neoplasias/terapia , Tolerância Imunológica , Imunoterapia , Inflamação
9.
Mol Biol Rep ; 51(1): 571, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38662155

RESUMO

Leukemia is a malignancy of the bone marrow and blood originating from self-renewing cancerous immature blast cells or transformed leukocytes. Despite improvements in treatments, leukemia remains still a serious disease with poor prognosis because of disease heterogeneity, drug resistance and relapse. There is emerging evidence that differentially expression of co-signaling molecules play a critical role in tumor immune evasion. Galectin-9 (Gal-9) is one of the key proteins that leukemic cells express, secrete, and use to proliferate, self-renew, and survive. It also suppresses host immune responses controlled by T and NK cells, enabling leukemic cells to evade immune surveillance. The present review provides the molecular mechanisms of Gal-9-induced immune evasion in leukemia. Understanding the complex immune evasion machinery driven by Gal-9 expressing leukemic cells will enable the identification of novel therapeutic strategies for efficient immunotherapy in leukemic patients. Combined treatment approaches targeting T-cell immunoglobulin and mucin domain-3 (Tim-3)/Gal-9 and other immune checkpoint pathways can be considered, which may enhance the efficacy of host effector cells to attack leukemic cells.


Assuntos
Transformação Celular Neoplásica , Galectinas , Receptor Celular 2 do Vírus da Hepatite A , Leucemia , Humanos , Galectinas/metabolismo , Leucemia/imunologia , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/genética , Animais , Tolerância Imunológica , Transdução de Sinais , Evasão Tumoral , Proliferação de Células , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo
10.
Theranostics ; 14(6): 2589-2604, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38646647

RESUMO

Background: The mechanisms underlying the increased mortality of secondary infections during the immunosuppressive phase of sepsis remain elusive. Objectives: We sought to investigate the role of Siglec-F+ neutrophils on splenic T lymphocytes in the immunosuppressed phase of sepsis and on secondary infection in PICS mice, and to elucidate the underlying mechanisms. Methods: We established a mouse model of sepsis-induced immunosuppression followed by secondary infection with LPS or E. coli. The main manifestation of immunosuppression is the functional exhaustion of splenic T lymphocytes. Treg depletion reagent Anti-IL-2, IL-10 blocker Anti-IL-10R, macrophage depletion reagent Liposomes, neutrophil depletion reagent Anti-Ly6G, neutrophil migration inhibitor SB225002, Siglec-F depletion reagent Anti-Siglec-F are all used on PICS mice. The function of neutrophil subsets was investigated by adoptive transplantation and the experiments in vitro. Results: Compared to other organs, we observed a significant reduction in pro-inflammatory cytokines in the spleen, accompanied by a marked increase in IL-10 production, primarily by infiltrating neutrophils. These infiltrating neutrophils in the spleen during the immunosuppressive phase of sepsis undergo phenotypic change in the local microenvironment, exhibiting high expression of neutrophil biomarkers such as Siglec-F, Ly6G, and Siglec-E. Depletion of neutrophils or specifically targeting Siglec-F leads to enhance the function of T lymphocytes and a notable improvement in the survival of mice with secondary infections. Conclusions: We identified Siglec-F+ neutrophils as the primary producers of IL-10, which significantly contributed to T lymphocyte suppression represents a novel finding with potential therapeutic implications.


Assuntos
Interleucina-10 , Neutrófilos , Sepse , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico , Baço , Animais , Baço/imunologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Camundongos , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/metabolismo , Sepse/imunologia , Interleucina-10/metabolismo , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Terapia de Imunossupressão , Infecções por Escherichia coli/imunologia , Masculino , Citocinas/metabolismo , Tolerância Imunológica , Linfócitos T Reguladores/imunologia
11.
Front Immunol ; 15: 1339714, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38571951

RESUMO

The intricate balance of immune reactions towards invading pathogens and immune tolerance towards self is pivotal in preventing autoimmune diseases, with the thymus playing a central role in establishing and maintaining this equilibrium. The induction of central immune tolerance in the thymus involves the elimination of self-reactive T cells, a mechanism essential for averting autoimmunity. Disruption of the thymic T cell selection mechanisms can lead to the development of autoimmune diseases. In the dynamic microenvironment of the thymus, T cell migration and interactions with thymic stromal cells are critical for the selection processes that ensure self-tolerance. Thymic epithelial cells are particularly significant in this context, presenting self-antigens and inducing the negative selection of autoreactive T cells. Further, the synergistic roles of thymic fibroblasts, B cells, and dendritic cells in antigen presentation, selection and the development of regulatory T cells are pivotal in maintaining immune responses tightly regulated. This review article collates these insights, offering a comprehensive examination of the multifaceted role of thymic tissue homeostasis in the establishment of immune tolerance and its implications in the prevention of autoimmune diseases. Additionally, the developmental pathways of the thymus are explored, highlighting how genetic aberrations can disrupt thymic architecture and function, leading to autoimmune conditions. The impact of infections on immune tolerance is another critical area, with pathogens potentially triggering autoimmunity by altering thymic homeostasis. Overall, this review underscores the integral role of thymic tissue homeostasis in the prevention of autoimmune diseases, discussing insights into potential therapeutic strategies and examining putative avenues for future research on developing thymic-based therapies in treating and preventing autoimmune conditions.


Assuntos
Doenças Autoimunes , Timo , Humanos , Tolerância Imunológica , Doenças Autoimunes/prevenção & controle , Tolerância a Antígenos Próprios , Homeostase
12.
Trials ; 25(1): 272, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38641837

RESUMO

BACKGROUND: Children with sensitization against foods have to be orally food-challenged before eating these foods for the first time. However, the waiting time for an oral food challenge (OFC) in Germany is about 3-6 months. In contrast, there are hints that an early introduction of allergenic foods might be protective regarding the development of food allergy. The aim of this clinical trial is therefore to investigate, whether an introduction and regular consumption of small amounts of food allergens is safe and will result in an increase of tolerance in children with sensitization against food allergens with unknown clinical relevance. METHODS: In this randomized, placebo-controlled, double-blind, single-center trial, 138 children (8 months to 4 years of age) sensitized to the target allergen(s) hen's egg, cow's milk, peanuts, and/or hazelnuts with unknown clinical relevance will be randomized in a 1:1 ratio to either an active or a placebo group, daily receiving a rusk-like biscuit powder with or without the target allergen(s) for 3-6 months until an OFC will be performed in routine diagnostics. The primary endpoint is an IgE-mediated food allergy to the primary target allergen, after the interventional period. DISCUSSION: Children with sensitization against food allergens with unknown clinical relevance often have to avoid the corresponding foods for several months until an OFC is performed. Therefore, the "window of opportunity" for an early preventive introduction of allergenic foods might be missed. This trial will assess whether an introduction of small allergen amounts will favor tolerance development in these children. TRIAL REGISTRATION: German Clinical Trials Register DRKS00032769. Registered on 02 October 2023.


Assuntos
Galinhas , Hipersensibilidade Alimentar , Criança , Lactente , Bovinos , Humanos , Feminino , Animais , Hipersensibilidade Alimentar/diagnóstico , Hipersensibilidade Alimentar/prevenção & controle , Leite/efeitos adversos , Alérgenos/efeitos adversos , Tolerância Imunológica
13.
Adv Exp Med Biol ; 1444: 67-82, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38467973

RESUMO

Regulatory T (Treg) cells, which specifically express the master transcription factor FoxP3, are indispensable for the maintenance of immunological self-tolerance and homeostasis. Their functional or numerical anomalies can be causative of autoimmune and other inflammatory diseases. Recent advances in the research of the cellular and molecular basis of how Treg cells develop, exert suppression, and maintain their function have enabled devising various ways for controlling physiological and pathological immune responses by targeting Treg cells. It is now envisaged that Treg cells as a "living drug" are able to achieve antigen-specific immune suppression of various immune responses and reestablish immunological self-tolerance in the clinic.


Assuntos
Autoimunidade , Linfócitos T Reguladores , Tolerância a Antígenos Próprios , Tolerância Imunológica , Regulação da Expressão Gênica , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo
14.
Sci Transl Med ; 16(738): eadm8859, 2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38478632

RESUMO

Engineered regulatory T (Treg) cells have emerged as precision therapeutics aimed at inducing immune tolerance while reducing the risks associated with generalized immunosuppression. This Viewpoint highlights the opportunities and challenges for engineered Treg cell therapies in treating autoimmune and other inflammatory diseases.


Assuntos
Doenças Autoimunes , Linfócitos T Reguladores , Humanos , Tolerância Imunológica , Terapia de Imunossupressão
15.
Anaerobe ; 86: 102841, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38521227

RESUMO

OBJECTIVES: Clostridioides difficile infection (CDI) is the leading hospital-acquired infection in North America. While previous work on fecal microbiota transplantation (FMT), a highly effective treatment for CDI, has focused on colonization resistance mounted against C. difficile by FMT-delivered commensals, the effects of FMT on host gene expression are relatively unexplored. This study aims to identify transcriptional changes associated with FMT, particularly changes associated with protective immune responses. METHODS: Gene expression was assessed on day 2 and day 7 after FMT in mice after antibiotic-induced dysbiosis. Flow cytometry was also performed on colon and mesenteric lymph nodes at day 7 to investigate changes in immune cell populations. RESULTS: FMT administration after antibiotic-induced dysbiosis successfully restored microbial alpha diversity to levels of donor mice by day 7 post-FMT. Bulk RNA sequencing of cecal tissue at day 2 identified immune genes, including both pro-inflammatory and Type 2 immune pathways as upregulated after FMT. RNA sequencing was repeated on day 7 post-FMT, and expression of these immune genes was decreased along with upregulation of genes associated with restoration of intestinal homeostasis. Immunoprofiling on day 7 identified increased colonic CD45+ immune cells that exhibited dampened Type 1 and heightened regulatory and Type 2 responses. These include an increased abundance of eosinophils, alternatively activated macrophages, Th2, and T regulatory cell populations. CONCLUSION: These results highlight the impact of FMT on host gene expression, providing evidence that FMT restores intestinal homeostasis after antibiotic treatment and facilitates tolerogenic and Type 2 immune responses.


Assuntos
Infecções por Clostridium , Modelos Animais de Doenças , Transplante de Microbiota Fecal , Animais , Transplante de Microbiota Fecal/métodos , Camundongos , Infecções por Clostridium/terapia , Infecções por Clostridium/imunologia , Infecções por Clostridium/microbiologia , Microbioma Gastrointestinal , Disbiose/terapia , Clostridioides difficile/imunologia , Tolerância Imunológica , Camundongos Endogâmicos C57BL
16.
CNS Neurosci Ther ; 30(3): e14643, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38470096

RESUMO

AIMS: Glioblastoma is the most frequent and aggressive primary brain tumor, characterized by rapid disease course and poor treatment responsiveness. The abundance of immunosuppressive macrophages in glioblastoma challenges the efficacy of novel immunotherapy. METHODS: Bulk RNA-seq and single-cell RNA-seq of glioma patients from public databases were comprehensively analyzed to illustrate macrophage infiltration patterns and molecular characteristics of podoplanin (PDPN). Multiplexed fluorescence immunohistochemistry staining of PDPN, GFAP, CD68, and CD163 were performed in glioma tissue microarray. The impact of PDPN on macrophage immunosuppressive polarization was investigated using a co-culture system. Bone marrow-derived macrophages (BMDMs) and OT-II T cells isolated from BALB/c and OT-II mice respectively were co-cultured to determine T-cell adherence. Pathway alterations were probed through RNA sequencing and western blot analyses. RESULTS: Our findings demonstrated that PDPN is notably correlated with the expression of CD68 and CD163 in glioma tissues. Additionally, macrophages phagocytosing PDPN-containing EVs (EVsPDPN ) from GBM cells presented increased CD163 expression and augmented secretion of immunoregulatory cytokine (IL-6, IL-10, TNF-α, and TGF-ß1). PDPN within EVs was also associated with enhanced phagocytic activity and reduced MHC II expression in macrophages, compromising CD4+ T-cell activation. CONCLUSIONS: This investigation underscores that EVsPDPN derived from glioblastoma cells contributes to M2 macrophage-mediated immunosuppression and is a potential prognostic marker and therapeutic target in glioblastoma.


Assuntos
Exossomos , Glioblastoma , Glioma , Animais , Humanos , Camundongos , Exossomos/metabolismo , Glioblastoma/patologia , Glioma/metabolismo , Tolerância Imunológica , Fatores de Transcrição , Macrófagos Associados a Tumor/metabolismo , Macrófagos Associados a Tumor/patologia
17.
Int J Biol Macromol ; 264(Pt 2): 130621, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38447834

RESUMO

The immune system acts as a vital defense barrier against pathogenic invasions, and its stable operation is crucial for maintaining body health. Nevertheless, various natural or artificial factors can compromise the body's immune function, leading to immunosuppression, which may interfere with the efficacy of vaccination and increase the susceptibility of the body to disease-causing pathogens. In an effort to ensure successful vaccinations and improve overall physical well-being, the search for appropriate immune regulators to enhance immunity is of paramount importance. Lentinan (LNT) has a significant role in immune regulation and vaccine adjuvants. In the present study, we constructed an immunosuppressive model using dexamethasone (DEX) and demonstrated that LNT could significantly improved antibody levels in immunosuppressive mice and stimulated T-lymphocyte proliferation and differentiation in intestinal Peyer's patches. LNT also increased the production of secretory immunoglobulin A (sIgA) in the duodenal fluid, the number of goblet cells, and the proportion of mucin area. Moreover, LNT modulated the intestinal microbiota and increased the production of short-chain fatty acids. Additionally, LNT promoted the proliferation, differentiation, and pro-inflammatory cytokines production of DEX-treated splenic T lymphocytes in vitro. Thus, the present study highlights the potential of LNT in reversing immunosuppression and avoiding the failure of vaccination.


Assuntos
Terapia de Imunossupressão , Lentinano , Animais , Camundongos , Lentinano/farmacologia , Tolerância Imunológica , Intestinos , Dexametasona/farmacologia
18.
Int Immunopharmacol ; 131: 111806, 2024 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-38457985

RESUMO

Cow's milk protein allergy (CMPA) is primarily due to the inability of the intestinal mucosa to establish typical immunological tolerance to proteins found in cow's milk, and the specific molecular mechanism is still unclear. In order to investigate molecular alterations in intestinal tissues during CMPA occurrence, this study analyzed the jejunal tissue of ß-lactoglobulin (BLG)-sensitized mice through transcriptomics and quantitative tandem mass tag (TMT)-labeled proteomics. A total of 475 differentially expressed genes (256 up-regulated, 219 down-regulated) and 94 differentially expressed proteins (65 up-regulated, 29 down-regulated) were identified. Comparing the KEGG pathways of the two groups, it was found that both were markedly enriched in the signaling pathways of complement and coagulation cascade. Among these, kallikrein B1 (KLKB1) in this pathway is speculated to be pivotal in CMPA. It may potentially enhance the release of bradykinin by activating the kallikrein-kinin system, leading to pro-inflammatory effects and exacerbating intestinal mucosal damage. This study suggests that the pathways of complement and coagulation cascades could be significant in the context of intestinal immunity in CMPA, and KLKB1 may be its potential therapeutic target.


Assuntos
Hipersensibilidade a Leite , Bovinos , Feminino , Animais , Camundongos , Hipersensibilidade a Leite/genética , Proteômica , Leite , Perfilação da Expressão Gênica , Tolerância Imunológica
19.
Sci Immunol ; 9(93): eadj4775, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38489352

RESUMO

The gut microbiota promotes immune system development in early life, but the interactions between the gut metabolome and immune cells in the neonatal gut remain largely undefined. Here, we demonstrate that the neonatal gut is uniquely enriched with neurotransmitters, including serotonin, and that specific gut bacteria directly produce serotonin while down-regulating monoamine oxidase A to limit serotonin breakdown. We found that serotonin directly signals to T cells to increase intracellular indole-3-acetaldehdye and inhibit mTOR activation, thereby promoting the differentiation of regulatory T cells, both ex vivo and in vivo in the neonatal intestine. Oral gavage of serotonin into neonatal mice resulted in long-term T cell-mediated antigen-specific immune tolerance toward both dietary antigens and commensal bacteria. Together, our study has uncovered an important role for specific gut bacteria to increase serotonin availability in the neonatal gut and identified a function of gut serotonin in shaping T cell response to dietary antigens and commensal bacteria to promote immune tolerance in early life.


Assuntos
Microbioma Gastrointestinal , Serotonina , Animais , Camundongos , Bactérias , Tolerância Imunológica , Antígenos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...