Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.457
Filtrar
1.
Chem Biol Interact ; 395: 111011, 2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38653352

RESUMEN

Immune homeostasis is key to guarantee that the immune system can elicit effector functions against pathogens and at the same time raise tolerance towards other antigens. A disturbance of this delicate balance may underlie or at least trigger pathologies. Endocrine disrupting chemicals (EDCs) are increasingly recognized as risk factors for immune dysregulation. However, the immunotoxic potential of specific EDCs and their mixtures is still poorly understood. Thus, we aimed to investigate the effect of bisphenol A (BPA) and benzophenone-3 (BP-3), alone and in combination, on in vitro differentiation of T helper (TH)17 cells and regulatory T (Treg) cells. Naïve T cells were isolated from mouse lymphoid tissues and differentiated into the respective TH population in the presence of 0.001-10 µM BP-3 and/or 0.01-100 µM BPA. Cell viability, proliferation and the expression of TH lineage specific transcription factors and cytokines was measured by flow cytometry and CBA/ELISA. Moreover, the transcription of hormone receptors as direct targets of EDCs was quantified by RT-PCR. We found that the highest BPA concentration adversely affected TH cell viability and proliferation. Moreover, the general differentiation potential of both TH populations was not altered in the presence of both EDCs. However, EDC exposure modulated the emergence of TH17 and Treg cell intermediate states. While BPA and BP-3 promoted the development of TH1-like TH17 cells under TH17-differentiating conditions, TH2-like Treg cells occurred under Treg polarization. Interestingly, differential effects could be observed in mixtures of the two tested compounds compared with the individual compounds. Notably, estrogen receptor ß expression was decreased under TH17-differentiating conditions in the presence of BPA and BP-3 as mixture. In conclusion, our study provides solid evidence for both, the immune disruptive potential and the existence of cumulative effects of real nature EDC mixtures on T cell in vitro differentiation.


Asunto(s)
Compuestos de Bencidrilo , Benzofenonas , Diferenciación Celular , Fenoles , Linfocitos T Reguladores , Células Th17 , Fenoles/toxicidad , Fenoles/farmacología , Animales , Compuestos de Bencidrilo/toxicidad , Benzofenonas/farmacología , Benzofenonas/toxicidad , Diferenciación Celular/efectos de los fármacos , Ratones , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Células Th17/efectos de los fármacos , Células Th17/citología , Células Th17/metabolismo , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Proliferación Celular/efectos de los fármacos , Disruptores Endocrinos/toxicidad , Disruptores Endocrinos/farmacología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/citología , Células Cultivadas
2.
Proc Natl Acad Sci U S A ; 119(34): e2206208119, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-35969754

RESUMEN

Although glioblastoma multiforme (GBM) is not an invariably cold tumor, checkpoint inhibition has largely failed in GBM. In order to investigate T cell-intrinsic properties that contribute to the resistance of GBM to endogenous or therapeutically enhanced adaptive immune responses, we sorted CD4+ and CD8+ T cells from the peripheral blood, normal-appearing brain tissue, and tumor bed of nine treatment-naive patients with GBM. Bulk RNA sequencing of highly pure T cell populations from these different compartments was used to obtain deep transcriptomes of tumor-infiltrating T cells (TILs). While the transcriptome of CD8+ TILs suggested that they were partly locked in a dysfunctional state, CD4+ TILs showed a robust commitment to the type 17 T helper cell (TH17) lineage, which was corroborated by flow cytometry in four additional GBM cases. Therefore, our study illustrates that the brain tumor environment in GBM might instruct TH17 commitment of infiltrating T helper cells. Whether these properties of CD4+ TILs facilitate a tumor-promoting milieu and thus could be a target for adjuvant anti-TH17 cell interventions needs to be further investigated.


Asunto(s)
Neoplasias Encefálicas , Linfocitos T CD4-Positivos , Glioblastoma , Linfocitos T Colaboradores-Inductores , Neoplasias Encefálicas/patología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Citometría de Flujo , Glioblastoma/patología , Humanos , Linfocitos Infiltrantes de Tumor/citología , Linfocitos T Colaboradores-Inductores/citología
3.
J Cachexia Sarcopenia Muscle ; 13(4): 2088-2101, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35718758

RESUMEN

BACKGROUND: Cachexia is a life-threatening condition observed in several pathologies, such as cancer or chronic diseases. Interleukin 10 (Il10) gene transfer is known to improve cachexia by downregulating Il6. Here, we used an IL10-knockout mouse model to simulate cachexia and investigate the effects of eggshell membrane (ESM), a resistant protein, on general pre-cachexia symptoms, which is particularly important for the development of cachexia therapeutics. METHODS: Five-week-old male C57BL6/J mice were fed an AIN-93G powdered diet (WT), and 5-week-old male B6.129P2-Il10 < tm1Cgn>/J (IL10-/- ) mice were fed either the AIN-93G diet (KO) or an 8% ESM-containing diet (KOE) for 28 weeks. The tissue weight and levels of anaemia-, blood glucose-, lipid metabolism-, and muscular and colonic inflammation-related biochemical markers were measured. Transcriptomic analysis on liver and colon mucus and proteomic analysis on skeletal muscle were performed. Ingenuity Pathway Analysis was used to identify molecular pathways and networks. Caecal short-chain fatty acids (SCFAs) were identified using HPLC, and caecal bacteria DNA were subjected to metagenomic analysis. Flow cytometry analysis was performed to measure the CD4+ IL17+ T cells in mesenteric lymph nodes. RESULTS: The body weight, weight of gastrocnemius muscle and fat tissues, colon weight/length ratio, plasma HDL and NEFA, muscular PECAM-1 levels (P < 0.01), plasma glucose and colonic mucosal myeloperoxidase activity (P < 0.05) and T helper (Th) 17 cell abundance (P = 0.071) were improved in KOE mice over KO mice. Proteomic analysis indicated the protective role of ESM in muscle weakness and maintenance of muscle formation (>1.5-fold). Transcriptomic analysis revealed that ESM supplementation suppressed the LPS/IL1-mediated inhibition of RXR function pathway in the liver and downregulated the colonic mucosal expression of chemokines and Th cell differentiation-related markers (P < 0.01) by suppressing the upstream BATF pathway. Analysis of the intestinal microenvironment revealed that ESM supplementation ameliorated the microbial alpha diversity and the abundance of microbiota associated with the degree of inflammation (P < 0.05) and increased the level of total organic acids, particularly of SCFAs such as butyrate (2.3-fold), which could inhibit Th1 and Th17 production. CONCLUSIONS: ESM supplementation ameliorated the chief symptoms of cachexia, including anorexia, lean fat tissue mass, skeletal muscle wasting and reduced physical function. ESM also improved colon and skeletal muscle inflammation, lipid metabolism and microbial dysbiosis. These results along with the suppressed differentiation of Th cells could be associated with the beneficial intestinal microenvironment and, subsequently, attenuation of pre-cachexia. Our findings provide insights into the potential of ESM in complementary interventions for pre-cachexia prevention.


Asunto(s)
Caquexia , Cáscara de Huevo , Microbioma Gastrointestinal , Linfocitos T Colaboradores-Inductores , Animales , Caquexia/prevención & control , Diferenciación Celular , Dieta , Inflamación , Interleucina-10 , Masculino , Ratones , Ratones Endogámicos C57BL , Proteómica , Linfocitos T Colaboradores-Inductores/citología
4.
Cell Rep ; 38(8): 110399, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35139367

RESUMEN

Follicular helper T (Tfh) cells promote, whereas follicular regulatory T (Tfr) cells restrain, germinal center (GC) reactions. However, the precise roles of these cells in the complex GC reaction remain poorly understood. Here, we perturb Tfh or Tfr cells after SARS-CoV-2 spike protein vaccination in mice. We find that Tfh cells promote the frequency and somatic hypermutation (SHM) of Spike-specific GC B cells and regulate clonal diversity. Tfr cells similarly control SHM and clonal diversity in the GC but do so by limiting clonal competition. In addition, deletion of Tfh or Tfr cells during primary vaccination results in changes in SHM after vaccine boosting. Aged mice, which have altered Tfh and Tfr cells, have lower GC responses, presenting a bimodal distribution of SHM. Together, these data demonstrate that GC responses to SARS-CoV-2 spike protein vaccines require a fine balance of positive and negative follicular T cell help to optimize humoral immunity.


Asunto(s)
COVID-19/prevención & control , Centro Germinal/inmunología , Glicoproteína de la Espiga del Coronavirus/administración & dosificación , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Envejecimiento , Animales , Anticuerpos Antivirales/sangre , Linfocitos B/inmunología , Linfocitos B/metabolismo , COVID-19/virología , Centro Germinal/citología , Centro Germinal/metabolismo , Inmunidad Humoral , Ratones , Ratones Endogámicos C57BL , SARS-CoV-2/inmunología , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/inmunología , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Vacunación , Vacunas de Subunidad/inmunología
5.
Biochim Biophys Acta Mol Cell Res ; 1869(4): 119192, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34982961

RESUMEN

A Disintegrin and Metalloproteinases (ADAM)-10 is a member of a family of membrane-anchored proteinases that regulate a broad range of cellular functions with central roles within the immune system. This has spurred the interest to modulate ADAM activity therapeutically in immunological diseases. CD4 T helper (Th) cells are the key regulators of adaptive immune responses. Their development and function is strongly dependent on Notch, a key ADAM-10 substrate. However, Th cells rely on a variety of additional ADAM-10 substrates regulating their functional activity at multiple levels. The complexity of both, the ADAM substrate expression as well as the functional consequences of ADAM-mediated cleavage of the various substrates complicates the analysis of cell type specific effects. Here we provide an overview on the major ADAM-10 substrates relevant for CD4 T cell biology and discuss the potential effects of ADAM-mediated cleavage exemplified for a selection of important substrates.


Asunto(s)
Proteína ADAM10/metabolismo , Linfocitos T Colaboradores-Inductores/metabolismo , Proteína ADAM17/metabolismo , Antígeno B7-H1/metabolismo , Diferenciación Celular , Proliferación Celular , Humanos , Receptores de Interleucina-2/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología
6.
BMC Cancer ; 21(1): 1226, 2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-34781899

RESUMEN

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease with rising incidence and with 5-years overall survival of less than 8%. PDAC creates an immune-suppressive tumor microenvironment to escape immune-mediated eradication. Regulatory T (Treg) cells and myeloid-derived suppressor cells (MDSC) are critical components of the immune-suppressive tumor microenvironment. Shifting from tumor escape or tolerance to elimination is the major challenge in the treatment of PDAC. RESULTS: In a mathematical model, we combine distinct treatment modalities for PDAC, including 5-FU chemotherapy and anti- CD25 immunotherapy to improve clinical outcome and therapeutic efficacy. To address and optimize 5-FU and anti- CD25 treatment (to suppress MDSCs and Tregs, respectively) schedule in-silico and simultaneously unravel the processes driving therapeutic responses, we designed an in vivo calibrated mathematical model of tumor-immune system (TIS) interactions. We designed a user-friendly graphical user interface (GUI) unit which is configurable for treatment timings to implement an in-silico clinical trial to test different timings of both 5-FU and anti- CD25 therapies. By optimizing combination regimens, we improved treatment efficacy. In-silico assessment of 5-FU and anti- CD25 combination therapy for PDAC significantly showed better treatment outcomes when compared to 5-FU and anti- CD25 therapies separately. Due to imprecise, missing, or incomplete experimental data, the kinetic parameters of the TIS model are uncertain that this can be captured by the fuzzy theorem. We have predicted the uncertainty band of cell/cytokines dynamics based on the parametric uncertainty, and we have shown the effect of the treatments on the displacement of the uncertainty band of the cells/cytokines. We performed global sensitivity analysis methods to identify the most influential kinetic parameters and simulate the effect of the perturbation on kinetic parameters on the dynamics of cells/cytokines. CONCLUSION: Our findings outline a rational approach to therapy optimization with meaningful consequences for how we effectively design treatment schedules (timing) to maximize their success, and how we treat PDAC with combined 5-FU and anti- CD25 therapies. Our data revealed that a synergistic combinatorial regimen targeting the Tregs and MDSCs in both crisp and fuzzy settings of model parameters can lead to tumor eradication.


Asunto(s)
Carcinoma Ductal Pancreático/terapia , Fluorouracilo/uso terapéutico , Inmunoterapia/métodos , Subunidad alfa del Receptor de Interleucina-2/inmunología , Modelos Teóricos , Neoplasias Pancreáticas/terapia , Animales , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Lógica Difusa , Humanos , Tolerancia Inmunológica , Inmunidad Celular , Células Asesinas Naturales/citología , Ratones , Ratones Endogámicos C57BL , Células Supresoras de Origen Mieloide/efectos de los fármacos , Trasplante de Neoplasias , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Linfocitos T Citotóxicos/citología , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Reguladores/efectos de los fármacos , Resultado del Tratamiento , Escape del Tumor , Microambiente Tumoral/inmunología , Interfaz Usuario-Computador
7.
Cell Rep ; 36(11): 109696, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34525366

RESUMEN

CD4 T cells are essential for immunity to tuberculosis because they produce cytokines, including interferon-γ. Whether CD4 T cells act as "helper" cells to promote optimal CD8 T cell responses during Mycobacterium tuberculosis is unknown. Using two independent models, we show that CD4 T cell help enhances CD8 effector functions and prevents CD8 T cell exhaustion. We demonstrate synergy between CD4 and CD8 T cells in promoting the survival of infected mice. Purified helped, but not helpless, CD8 T cells efficiently restrict intracellular bacterial growth in vitro. Thus, CD4 T cell help plays an essential role in generating protective CD8 T cell responses against M. tuberculosis infection in vitro and in vivo. We infer vaccines that elicit both CD4 and CD8 T cells are more likely to be successful than vaccines that elicit only CD4 or CD8 T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Tuberculosis/inmunología , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Modelos Animales de Enfermedad , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium tuberculosis/patogenicidad , Mycobacterium tuberculosis/fisiología , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Tasa de Supervivencia , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/metabolismo , Transcriptoma , Tuberculosis/mortalidad , Tuberculosis/patología
8.
Vet Immunol Immunopathol ; 240: 110316, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34474261

RESUMEN

CD4+ helper T cells play key and diverse roles in inducing adaptive immune responses in vertebrates. The CD4 molecule, which is found on the surfaces of CD4+ helper T cells, can be used to distinguish subsets of helper T cells. Teleosts are the oldest living species with bona-fide CD4 coreceptors. Although some components of immune systems of teleosts and mammals appear to be similar, many physiological differences are represented between them. Previous studies have shown that two CD4 paralogs are present in teleosts, whereas only one is present in mammals. Therefore, in this review, the CD4 molecular structure, expression profiles, subpopulations, and biological functions of teleost CD4+ helper T cells were summarized and compared with those of their mammalian counterparts to understand the differences in CD4 molecules between teleosts and mammals. This review provides suggestions for further studies on the CD4 molecular function and regulatory mechanism of CD4+ helper T cells in teleost fish and will help establish therapeutic strategies to control fish diseases in the future.


Asunto(s)
Peces/inmunología , Linfocitos T Colaboradores-Inductores , Animales , Mamíferos/inmunología , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología
9.
PLoS One ; 16(7): e0252805, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34197491

RESUMEN

Chemokines are small proteins that promote leukocyte migration during development, infection, and inflammation. We and others isolated the unique chemokine CCL21, a potent chemo-attractant for naïve T-cells, naïve B-cells, and immature dendritic cells. CCL21 has a 37 amino acid carboxy terminal extension that is distinct from the rest of the chemokine family, which is thought to anchor it to venule endothelium where the amino terminus can interact with its cognate receptor, CCR7. We and others have reported that venule endothelium expressing CCL21 plays a crucial role in attracting naïve immune cells to sites of antigen presentation. In this study we generated a series of monoclonal antibodies to the amino terminus of CCL21 in an attempt to generate an antibody that blocked the interaction of CCL21 with its receptor CCR7. We found one humanized clone that blocked naïve T-cell migration towards CCL21, while memory effector T-cells were less affected. Using this monoclonal antibody, we also demonstrated that CCL21 is expressed in the mucosal venule endothelium of the large majority of inflammatory bowel diseases (IBD), including Crohn's disease, ulcerative colitis, and also in celiac disease. This expression correlated with active IBD in 5 of 6 cases, whereas none of 6 normal bowel biopsies had CCL21 expression. This study raises the possibility that this monoclonal antibody could be used to diagnose initial or recurrent of IBD. Significantly, this antibody could also be used for therapeutic intervention in IBD by selectively interfering with recruitment of naïve immune effector cells to sites of antigen presentation, without harming overall memory immunity.


Asunto(s)
Anticuerpos Monoclonales Humanizados/inmunología , Quimiocina CCL21/inmunología , Enfermedades Inflamatorias del Intestino/diagnóstico , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Quimiocina CCL21/genética , Quimiocina CCL21/metabolismo , Quimiotaxis/efectos de los fármacos , Endotelio/metabolismo , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/metabolismo , Ratones , Ratones Endogámicos BALB C , Unión Proteica , Receptores CCR7/metabolismo , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
10.
Cell ; 184(16): 4186-4202.e20, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34216540

RESUMEN

Polyamine synthesis represents one of the most profound metabolic changes during T cell activation, but the biological implications of this are scarcely known. Here, we show that polyamine metabolism is a fundamental process governing the ability of CD4+ helper T cells (TH) to polarize into different functional fates. Deficiency in ornithine decarboxylase, a crucial enzyme for polyamine synthesis, results in a severe failure of CD4+ T cells to adopt correct subset specification, underscored by ectopic expression of multiple cytokines and lineage-defining transcription factors across TH cell subsets. Polyamines control TH differentiation by providing substrates for deoxyhypusine synthase, which synthesizes the amino acid hypusine, and mice in which T cells are deficient for hypusine develop severe intestinal inflammatory disease. Polyamine-hypusine deficiency caused widespread epigenetic remodeling driven by alterations in histone acetylation and a re-wired tricarboxylic acid (TCA) cycle. Thus, polyamine metabolism is critical for maintaining the epigenome to focus TH cell subset fidelity.


Asunto(s)
Linaje de la Célula , Poliaminas/metabolismo , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cromatina/metabolismo , Ciclo del Ácido Cítrico/efectos de los fármacos , Colitis/inmunología , Colitis/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Epigenoma , Histonas/metabolismo , Inflamación/inmunología , Inflamación/patología , Subgrupos Linfocitarios/efectos de los fármacos , Subgrupos Linfocitarios/metabolismo , Lisina/análogos & derivados , Lisina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ornitina Descarboxilasa/metabolismo , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Células Th17/efectos de los fármacos , Células Th17/inmunología , Factores de Transcripción/metabolismo
11.
Nat Immunol ; 22(8): 969-982, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34312548

RESUMEN

The transcription factor ThPOK (encoded by the Zbtb7b gene) controls homeostasis and differentiation of mature helper T cells, while opposing their differentiation to CD4+ intraepithelial lymphocytes (IELs) in the intestinal mucosa. Thus CD4 IEL differentiation requires ThPOK transcriptional repression via reactivation of the ThPOK transcriptional silencer element (SilThPOK). In the present study, we describe a new autoregulatory loop whereby ThPOK binds to the SilThPOK to maintain its own long-term expression in CD4 T cells. Disruption of this loop in vivo prevents persistent ThPOK expression, leads to genome-wide changes in chromatin accessibility and derepresses the colonic regulatory T (Treg) cell gene expression signature. This promotes selective differentiation of naive CD4 T cells into GITRloPD-1loCD25lo (Triplelo) Treg cells and conversion to CD4+ IELs in the gut, thereby providing dominant protection from colitis. Hence, the ThPOK autoregulatory loop represents a key mechanism to physiologically control ThPOK expression and T cell differentiation in the gut, with potential therapeutic relevance.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Linfocitos Intraepiteliales/citología , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Reguladores/citología , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular/inmunología , Colitis/inmunología , Colitis/prevención & control , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Femenino , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Factores de Transcripción/genética , Transcripción Genética/genética
12.
Cells ; 10(5)2021 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-34066151

RESUMEN

High-affinity antibody-secreting cells (ASC) arise from terminal differentiation of B-cells after coordinated interactions with T follicular helper (Tfh) cells in germinal centers (GC). Elucidation of cues promoting human naive B-cells to progress into ASCs is challenging, as this process is notoriously difficult to induce in vitro while maintaining enough cell numbers to investigate the differentiation route(s). Here, we describe a minimalistic in vitro culture system that supports efficient differentiation of human naive B-cells into antibody-secreting cells. Upon initial stimulations, the interplay between level of CD40 costimulation and the Tfh cell-associated cytokines IL-21 and IL-4 determined the magnitude of B-cell expansion, immunoglobulin class-switching and expression of ASC regulator PRDM1. In contrast, the B-cell-specific transcriptional program was maintained, and efficient ASC formation was hampered. Renewed CD40 costimulation and Tfh cytokines exposure induced rapid secondary STAT3 signaling and extensive ASC differentiation, accompanied by repression of B-cell identity factors PAX5, BACH2 and IRF8 and further induction of PRDM1. Our work shows that, like in vivo, renewed CD40L costimulation also induces efficient terminal ASC differentiation after initial B-cell expansion in vitro. This culture system for efficient differentiation of human naive B-cells into ASCs, while also maintaining high cell numbers, may form an important tool in dissecting human naive B-cell differentiation, thereby enabling identification of novel transcriptional regulators and biomarkers for desired and detrimental antibody formation in humans.


Asunto(s)
Anticuerpos/química , Linfocitos B/citología , Células 3T3 , Animales , Formación de Anticuerpos , Antígenos CD40/metabolismo , Diferenciación Celular , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica , Centro Germinal/inmunología , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Técnicas In Vitro , Activación de Linfocitos/inmunología , Ratones , Células 3T3 NIH , Fosforilación , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Transducción de Señal , Linfocitos T Colaboradores-Inductores/citología
14.
Cell Mol Biol Lett ; 26(1): 7, 2021 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-33622250

RESUMEN

BACKGROUND: NOTCH signaling has been shown to play a role in the production of interleukin-22 (IL-22) by CD4+ T cells. Multiple T-helper (Th) cell populations secrete IL-22. Th22 (CD4+IL22+IFNγ-IL17A-) cells are a subgroup of CD4+ effector T cells that primarily generate IL-22. The regulatory mechanisms of the NOTCH signaling pathway involved in differentiation of the Th22 cell subset have not been completely elucidated. This study aimed to further explore the involvement of NOTCH signaling in Th22 differentiation. METHODS: In vitro combination of IL-6, IL-23, and tumor necrosis factor-α (TNF-α) treatment with naïve CD4+ T cells established the Th22 cell induced model. NOTCH signaling was activated by jagged-1 and inhibited by (2S)-N-[(3,5-difluorophenyl) acetyl]-L-alanyl-2-phenyl]glycine 1,1-dimethylethyl ester (DAPT). HES-1 siRNA and HES-1 vector were employed to knock down and induce overexpression of HES-1 to investigate the effect of NOTCH signaling on the differentiation of CD4+T cells into Th22 cells. RESULTS: We observed that the proportion of Th22 cells, along with Hes-1, Ahr, and Il-22 mRNA and protein expression, was increased by both jagged-1 and overexpression of HES-1. On the other hand, after the combined cytokine treatment of cells, and exposure to jagged-1 and DAPT or HES-1 siRNA, there was a decrease in the Th22 cell proportion, mRNA and protein expression of HES-1, AHR, and IL-22. CONCLUSIONS: Our study demonstrates that HES-1 enhancement in AHR and IL-22 up-regulation of NOTCH signaling can promote the skewing of naïve CD4+T cells toward Th22 cells. Also, the results of our study show that HES-1 is a crucial factor in Th22 cell differentiation.


Asunto(s)
Diferenciación Celular/inmunología , Interleucinas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Linfocitos T Colaboradores-Inductores/citología , Factor de Transcripción HES-1/metabolismo , Animales , Polaridad Celular/inmunología , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Interleucina-22
15.
Nature ; 592(7852): 133-137, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33597749

RESUMEN

Antibody affinity maturation depends on positive selection in germinal centres (GCs) of rare B cell clones that acquire higher-affinity B cell receptors via somatic hypermutation, present more antigen to follicular helper T (TFH) cells and, consequently, receive more contact-dependent T cell help1. As these GC B cells and TFH cells do not maintain long-lasting contacts in the chaotic GC environment2-4, it is unclear how sufficient T cell help is cumulatively focused onto those rare clones. Here we show that, upon stimulation of CD40, GC B cells upregulate the chemokine CCL22 and to a lesser extent CCL17. By engaging the chemokine receptor CCR4 on TFH cells, CCL22 and CCL17 can attract multiple helper cells from a distance, thus increasing the chance of productive help. During a GC response, B cells that acquire higher antigen-binding affinities express higher levels of CCL22, which in turn 'highlight' these high-affinity GC B cells. Acute increase or blockade of TFH cells helps to rapidly increase or decrease CCL22 expression by GC B cells, respectively. Therefore, a chemokine-based intercellular reaction circuit links the amount of T cell help that individual B cells have received recently to their subsequent ability to attract more help. When CCL22 and CCL17 are ablated in B cells, GCs form but B cells are not affinity-matured efficiently. When competing with wild-type B cells in the same reaction, B cells lacking CCL22 and CCL17 receive less T cell help to maintain GC participation or develop into bone-marrow plasma cells. By uncovering a chemokine-mediated mechanism that highlights affinity-improved B cells for preferential help from TFH cells, our study reveals a principle of spatiotemporal orchestration of GC positive selection.


Asunto(s)
Quimiocina CCL22/metabolismo , Centro Germinal/citología , Centro Germinal/inmunología , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Células Cultivadas , Quimiocina CCL17/deficiencia , Quimiocina CCL17/genética , Quimiocina CCL22/deficiencia , Quimiocina CCL22/genética , Femenino , Humanos , Masculino , Ratones , Tonsila Palatina/citología , Receptores CCR4/deficiencia , Receptores CCR4/genética , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología , Regulación hacia Arriba
16.
Nat Commun ; 12(1): 525, 2021 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-33483505

RESUMEN

CTLA-4 is an important regulator of T-cell function. Here, we report that expression of this immune-regulator in mouse B-1a cells has a critical function in maintaining self-tolerance by regulating these early-developing B cells that express a repertoire enriched for auto-reactivity. Selective deletion of CTLA-4 from B cells results in mice that spontaneously develop autoantibodies, T follicular helper (Tfh) cells and germinal centers (GCs) in the spleen, and autoimmune pathology later in life. This impaired immune homeostasis results from B-1a cell dysfunction upon loss of CTLA-4. Therefore, CTLA-4-deficient B-1a cells up-regulate epigenetic and transcriptional activation programs and show increased self-replenishment. These activated cells further internalize surface IgM, differentiate into antigen-presenting cells and, when reconstituted in normal IgH-allotype congenic recipient mice, induce GCs and Tfh cells expressing a highly selected repertoire. These findings show that CTLA-4 regulation of B-1a cells is a crucial immune-regulatory mechanism.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Antígeno CTLA-4/inmunología , Homeostasis/inmunología , Sistema Inmunológico/inmunología , Tolerancia Inmunológica/inmunología , Animales , Subgrupos de Linfocitos B/metabolismo , Antígeno CTLA-4/genética , Antígeno CTLA-4/metabolismo , Diferenciación Celular/inmunología , Centro Germinal/citología , Centro Germinal/inmunología , Centro Germinal/metabolismo , Sistema Inmunológico/citología , Sistema Inmunológico/metabolismo , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
17.
Rheumatology (Oxford) ; 60(1): 224-230, 2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-32671409

RESUMEN

OBJECTIVES: Influenza vaccination is effective in preventing infections in most people. This study aimed to assess the changes of immune responses in primary Sjögren's Syndrome (pSS) patients after influenza vaccination and determine the safety of influenza vaccination. METHODS: A total of 17 patients with pSS and 16 healthy controls (HCs) were included. Peripheral mononuclear cells were analysed by flow cytometry. Vaccine-specific antibodies were determined by ELISA. Clinical features and serological responses were monitored. RESULTS: The percentages of T follicular helper cell (Tfh) were significantly elevated in HCs after vaccination (P=0.0005), while no significant differences in the levels of Tfh in pSS patients were identified (P=0.1748). The proportions of Th2 cells were significantly decreased after vaccination in both pSS patients and HCs (P<0.05). In contrast, the percentages of Th1 cells and Th17 cells were significantly increased after vaccination in pSS patients (P<0.05), while no significant differences in the percentages of Th1 and Th17 cells were identified in HCs (P>0.05), although a trend towards higher levels of Th1 cells was observed (P=0.0830). No significant changes in the proportions of memory B cells and plasmablasts were observed after vaccination. Patients with pSS developed higher levels of vaccine-specific IgGs compared with HCs (P=0.001). No significant changes in disease manifestations and laboratory parameters were observed after vaccination. No increased vaccination related adverse effect was observed in pSS. CONCLUSION: Our findings suggest the feasibility of applying influenza vaccines to patients with pSS, raising awareness for vaccination among the rheumatology community and involved healthcare professionals.


Asunto(s)
Vacunas contra la Influenza/inmunología , Síndrome de Sjögren/inmunología , Linfocitos T Colaboradores-Inductores/citología , Adulto , Linfocitos B/citología , Estudios de Casos y Controles , Femenino , Citometría de Flujo , Humanos , Inmunidad Celular , Inmunoglobulina G/sangre , Vacunas contra la Influenza/administración & dosificación , Leucocitos Mononucleares/citología , Masculino , Persona de Mediana Edad , Células TH1/citología , Células Th17/citología , Vacunación
18.
Mol Cell Biochem ; 476(1): 369-384, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32965596

RESUMEN

There is limited information regarding the TLR2 signaling pathway involved in Th9 cell differentiation. The role of calcitriol in regulating TLR2-mediated Th9 cell development is unknown. Thus, we aimed to unravel the TLR2 signaling pathway in Th9 cells and its regulation by calcitriol. We have used n = 5-6 animals for each murine experiment. Human studies involved five healthy volunteers. Moreover, ten healthy individuals and ten RA patients were included in the study. Murine and human Th9 cells were treated with Calcitriol (100 nM) and Pam3CSK4 (2 µg/mL). The number of IL-9+ve cells was determined by flow cytometry. Real-time PCR was used to assess the gene expression. Serum 25(OH)D3 levels were determined by HPLC. We observed that TLR2 signals via IL-33/ST2 in Th9 cells. Increased TLR2 expression associated with increased IL9 expression and augmented disease severity in RA patients. Calcitriol attenuated TLR2 signaling in murine and human Th9 cells. Low serum vitamin D3 level negatively associated with increased IL-9 and TLR2 expression and disease severity in RA patients. Our data suggest a potential role of calcitriol to ameliorate the disease severity of RA patients.


Asunto(s)
Artritis Reumatoide/metabolismo , Calcitriol/farmacología , Interleucina-33/metabolismo , Receptor Toll-Like 2/metabolismo , Adulto , Animales , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular , Proliferación Celular , Femenino , Humanos , Interleucina-9/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Linfocitos T Colaboradores-Inductores/citología , Factores de Transcripción
19.
Scand J Immunol ; 93(5): e13008, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33314191

RESUMEN

In clinical practice, the capacity for producing anti-carbohydrate antibodies is regarded as an entity, but supportive evidence is lacking. We hypothesized that the outcome of the gold standard test for clinical assessment of this capacity, antibody response to polysaccharide vaccination, correlated with the level of the abundant naturally occurring anti-carbohydrate antibody, anti-αGal. To perform an exploratory study, 47 HIV-infected adults were recruited from a vaccine trial. Participants received a 23-valent pneumococcal capsular polysaccharide vaccine. Plasma samples obtained just before and median 4 weeks after the vaccination were quantified for IgG anti-αGal antibody and IgG antibodies to polysaccharides present in the vaccine (serotypes 1, 7F and 19A) by solid-phase type immunoassays. The vaccination responses were assessed as a categorical variable (based on criteria defined by The American Academy of Allergy, Asthma & Immunology and the American College of Allergy, Asthma & Immunology) and as three different continuous variables (antibody increment, geometrical average and standard normal deviates of the achieved antibody concentrations). The baseline anti-αGal level predicted the vaccine response as a categorical variable (ROC-curve analysis, AUC = 0.71; 95%CI: 0.55-0.86) and as the three continuous variables (eg slope of linear regression of geometrical average = 0.37; 95%CI: 0.15-0.59). The correlation between the anti-αGal level and antibody responses to polysaccharide vaccination fits with a shared underlying capacity. Thus, the present study supports the notion of a measurable capacity for the production of anti-carbohydrate antibodies in each individual. Firm conclusions on the generalizability and clinical utility require further studies.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Infecciones por VIH/inmunología , Vacunas Neumococicas/inmunología , Polisacáridos Bacterianos/inmunología , alfa-Galactosidasa/inmunología , Anticuerpos Antibacterianos/inmunología , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología
20.
J Virol ; 95(4)2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33208449

RESUMEN

This report evaluates a dietary manipulation approach to suppress the severity of ocular infections caused by herpes simplex virus infection. The virus causes chronic damage to the cornea that results from a T-cell-orchestrated inflammatory reaction to the infection. Lesion severity can be limited if cells with regulatory activity predominate over proinflammatory T cells and nonlymphoid inflammatory cells. In this report, we show that this outcome can be achieved by including the short-chain fatty acid (SCFA) salt sodium propionate (SP) in the drinking water. Animals given the SP supplement developed significantly fewer ocular lesions than those receiving no supplement. Corneas and lymphoid organs contained fewer CD4 Th1 and Th17 T cells, neutrophils, and macrophages than those of controls, but a higher frequency of regulatory T cells (Treg) was present. The inclusion of SP in cultures to induce CD4 T cell subsets in vitro reduced the magnitude of Th1 and Th17 responses but expanded Treg induction. Dietary manipulation was an effective approach to limit the severity of viral immuno-inflammatory lesions and may be worth exploring as a means to reduce the impact of herpetic lesions in humans.IMPORTANCE Herpetic lesions are a significant problem, and they are difficult to control with therapeutics. Our studies show that the severity of herpetic lesions in a mouse model can be diminished by changing the diet to include increased levels of SCFA, which act to inhibit the involvement of inflammatory T cells. We suggest that changing the diet to include higher levels of SCFA might be a useful approach to reducing the impact of recurrent herpetic lesions in humans.


Asunto(s)
Córnea , Suplementos Dietéticos , Ácidos Grasos Volátiles/administración & dosificación , Queratitis Herpética/dietoterapia , Propionatos/administración & dosificación , Animales , Células Cultivadas , Córnea/inmunología , Córnea/virología , Herpesvirus Humano 1/inmunología , Queratitis Herpética/inmunología , Queratitis Herpética/virología , Macrófagos/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/citología , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Reguladores/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...