Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28.833
Filtrar
Más filtros

Intervalo de año de publicación
1.
Biomaterials ; 312: 122750, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39126779

RESUMEN

Infiltration of immunosuppressive cells into the breast tumor microenvironment (TME) is associated with suppressed effector T cell (Teff) responses, accelerated tumor growth, and poor clinical outcomes. Previous studies from our group and others identified infiltration of immunosuppressive myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) as critical contributors to immune dysfunction in the orthotopic claudin-low tumor model, limiting the efficacy of adoptive cellular therapy. However, approaches to target these cells in the TME are currently lacking. To overcome this barrier, polymeric micellular nanoparticles (PMNPs) were used for the co-delivery of small molecule drugs activating Toll-like receptors 7 and 8 (TLR7/8) and inhibiting PI3K delta (PI3Kδ). The immunomodulation of the TME by TLR7/8 agonist and PI3K inhibitor led to type 1 macrophage polarization, decreased MDSC accumulation and selectively decreased tissue-resident Tregs in the TME, while enhancing the T and B cell adaptive immune responses. PMNPs significantly enhanced the anti-tumor activity of local radiation therapy (RT) in mice bearing orthotopic claudin-low tumors compared to RT alone. Taken together, these data demonstrate that RT combined with a nanoformulated immunostimulant diminished the immunosuppressive TME resulting in tumor regression. These findings set the stage for clinical studies of this approach.


Asunto(s)
Nanopartículas , Receptor Toll-Like 7 , Receptor Toll-Like 8 , Microambiente Tumoral , Animales , Microambiente Tumoral/efectos de los fármacos , Receptor Toll-Like 7/agonistas , Femenino , Nanopartículas/química , Ratones , Receptor Toll-Like 8/agonistas , Inmunomodulación/efectos de los fármacos , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I , Células Supresoras de Origen Mieloide/efectos de los fármacos , Células Supresoras de Origen Mieloide/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Ratones Endogámicos BALB C , Micelas , Humanos
2.
Methods Mol Biol ; 2857: 79-87, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39348056

RESUMEN

Rheumatoid arthritis (RA) is linked to various signs of advanced aging, such as premature immunosenescence which occurs due to decline in regenerative ability of T cells. RA T cells develop a unique aggressive inflammatory senescent phenotype with an imbalance of Th17/T regulatory (Treg) cell homeostasis and presence of CD28- T cells. The phenotypic analysis and characterization of T cell subsets become necessary to ascertain if any functional deficiencies exist within with the help of transcription factor (TF) analysis. These subset-specific TFs dictate the functional characteristics of T-cell populations, leading to the production of distinct effector cytokines and functions. Examining the expression, activity, regulation, and genetic sequence of TFs not only aids researchers in determining their importance in disease processes but also aids in immunological monitoring of patients enrolled in clinical trials, particularly in evaluating various T-cell subsets [Th17 (CD3+CD4+IL17+RORγt+) cells and T regulatory (Treg) (CD3+CD4+CD25+CD127-FOXP3+) cells], markers of T-cell aging [aged Th17 cells (CD3+CD4+IL17+RORγt+CD28-), and aged Treg cells (CD3+CD4+CD25+CD127-FOXP3+CD28-)]. In this context, we propose and outline the protocols for assessing the expression of TFs in aged Th17 and Treg cells, highlighting the crucial aspects of this cytometric approach.


Asunto(s)
Artritis Reumatoide , Inmunosenescencia , Linfocitos T Reguladores , Factores de Transcripción , Humanos , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Artritis Reumatoide/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Citometría de Flujo/métodos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Biomarcadores
3.
Nat Commun ; 15(1): 9143, 2024 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-39443450

RESUMEN

Dendritic cells (DCs) are crucial for initiating protective immune responses and have also been implicated in the generation and regulation of Foxp3+ regulatory T cells (Treg cells). Here, we show that in the lamina propria of the small intestine, the alternative NF-κB family member RelB is necessary for the differentiation of cryptopatch and isolated lymphoid follicle-associated DCs (CIA-DCs). Moreover, single-cell RNA sequencing reveals a RelB-dependent signature in migratory DCs in mesenteric lymph nodes favoring DC-Treg cell interaction including elevated expression and release of the chemokine CCL22 from RelB-deficient conventional DCs (cDCs). In line with the key role of CCL22 to facilitate DC-Treg cell interaction, RelB-deficient DCs have a selective advantage to interact with Treg cells in an antigen-specific manner. In addition, DC-specific RelB knockout animals show increased total Foxp3+ Treg cell numbers irrespective of inflammatory status. Consequently, DC-specific RelB knockout animals fail to mount protective Th2-dominated immune responses in the intestine after infection with Heligmosomoides polygyrus bakeri. Thus, RelB expression in cDCs acts as a rheostat to establish a tolerogenic set point that is maintained even during strong type 2 immune conditions and thereby is a key regulator of intestinal homeostasis.


Asunto(s)
Células Dendríticas , Tolerancia Inmunológica , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Reguladores , Factor de Transcripción ReIB , Animales , Factor de Transcripción ReIB/metabolismo , Factor de Transcripción ReIB/genética , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Linfocitos T Reguladores/inmunología , Ratones , Movimiento Celular , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Quimiocina CCL22/metabolismo , Quimiocina CCL22/genética , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Intestino Delgado/inmunología , Intestino Delgado/metabolismo , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Diferenciación Celular , Células Th2/inmunología
4.
Sci Transl Med ; 16(769): eadj9331, 2024 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-39413160

RESUMEN

Regulatory T cells (Tregs) make major contributions to immune homeostasis. Because Treg dysfunction can lead to both allo- and autoimmunity, there is interest in correcting these disorders through Treg adoptive transfer. Two of the central challenges in clinically deploying Treg cellular therapies are ensuring phenotypic stability and maximizing potency. Here, we describe an approach to address both issues through the creation of OX40 ligand (OX40L)-specific chimeric antigen receptor (CAR)-Tregs under the control of a synthetic forkhead box P3 (FOXP3) promoter. The creation of these CAR-Tregs enabled selective Treg stimulation by engagement of OX40L, a key activation antigen in alloimmunity, including both graft-versus-host disease and solid organ transplant rejection, and autoimmunity, including rheumatoid arthritis, systemic sclerosis, and systemic lupus erythematosus. We demonstrated that OX40L-CAR-Tregs were robustly activated in the presence of OX40L-expressing cells, leading to up-regulation of Treg suppressive proteins without induction of proinflammatory cytokine production. Compared with control Tregs, OX40L-CAR-Tregs more potently suppressed alloreactive T cell proliferation in vitro and were directly inhibitory toward activated monocyte-derived dendritic cells (DCs). We identified trogocytosis as one of the central mechanisms by which these CAR-Tregs effectively decrease extracellular display of OX40L, resulting in decreased DC stimulatory capacity. OX40L-CAR-Tregs demonstrated an enhanced ability to control xenogeneic graft-versus-host disease compared with control Tregs without abolishing the graft-versus-leukemia effect. These results suggest that OX40L-CAR-Tregs may have wide applicability as a potent cellular therapy to control both allo- and autoimmune diseases.


Asunto(s)
Células Presentadoras de Antígenos , Ligando OX40 , Linfocitos T Reguladores , Humanos , Linfocitos T Reguladores/inmunología , Ligando OX40/metabolismo , Células Presentadoras de Antígenos/inmunología , Animales , Receptores Quiméricos de Antígenos/metabolismo , Proliferación Celular , Factores de Transcripción Forkhead/metabolismo , Enfermedad Injerto contra Huésped/inmunología , Activación de Linfocitos/inmunología , Ratones
5.
Gut Microbes ; 16(1): 2416517, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39411786

RESUMEN

Infecting humans with controlled doses of helminths, such as human hookworm (termed hookworm therapy), is proposed to prevent or treat various intestinal and extraintestinal diseases. However, full-scale clinical trials examining hookworm therapy are limited by the inability to scale-up the production of hookworm larvae to infect sufficient numbers of patients. With the aim of overcoming this challenge, this study infected four healthy individuals with hookworm larvae that had been reanimated from cryopreserved eggs to examine their viability and immunogenicity. We demonstrate that reanimated cryopreserved hookworm larvae establish a viable hookworm infection and elicit a similar immune response to larvae cultured from fresh stool. Furthermore, a refined understanding of the therapeutic mechanisms of hookworm is imperative to determine which diseases to target with hookworm therapy. To investigate potential therapeutic mechanisms, this study assessed changes in the immune cells, microbiome, and plasma metabolome in the four healthy individuals infected with cryopreserved hookworm larvae and another nine individuals infected with larvae cultured from freshly obtained stool. We identified potential immunoregulatory mechanisms by which hookworm may provide a beneficial effect on its host, including increased expression of CTLA-4 on regulatory T cells (Tregs) and upregulation of tryptophan metabolism. Furthermore, we found that a participant's baseline microbiome predicted the severity of symptoms and intestinal inflammation experienced during a controlled hookworm infection. In summary, our findings demonstrate the feasibility of full-scale clinical trials examining hookworm therapy by minimizing the reliance on human donors and optimizing the culturing process, thereby enabling viable hookworm larvae to be mass-produced and enabling on-demand inoculation of patients. Furthermore, this study provides insights into the complex interactions between helminths and their host, which could inform the development of novel therapeutic strategies.


Asunto(s)
Antígeno CTLA-4 , Criopreservación , Infecciones por Uncinaria , Larva , Linfocitos T Reguladores , Triptófano , Humanos , Linfocitos T Reguladores/inmunología , Triptófano/metabolismo , Animales , Antígeno CTLA-4/metabolismo , Antígeno CTLA-4/genética , Infecciones por Uncinaria/inmunología , Infecciones por Uncinaria/parasitología , Larva/inmunología , Regulación hacia Arriba , Adulto , Femenino , Heces/parasitología , Heces/microbiología , Ancylostomatoidea/inmunología , Masculino
7.
BMC Immunol ; 25(1): 68, 2024 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-39402453

RESUMEN

We previously demonstrated that the overall number of regulatory T (Treg) cells decrease proportionately with helper CD4+ T cells and their frequencies increase in antiretroviral therapy (ART)-naive human immunodeficiency virus type-1 (HIV-1) infected individuals. The question now is whether the discrepancies in Treg cell numbers and frequencies are synonymous to an impairment of their functions. To address this, we purified Treg cells and assessed their ability to modulate autologous monocytes functions. We observed that Treg cells were able to down modulate autologous monocytes activation as well as interleukin 6 (IL-6) and tumor necrosis factor-alpha (TNF-α) production during stimulation with polyinosinic-polycytidylic acid stabilized with poly-L-lysine and carboxymethylcellulose (poly-ICLC). This activity of Treg cells has been shown to be influenced by immunocompetence including but not limited to helper CD4+ T cell counts, in individuals with HIV-1 infection. Compared to immunosuppressed participants (CD4 < 500 cells/µL), immunocompetent participants (CD4 ≥ 500 cells/µL) showed significantly higher levels of transforming growth factor beta (TGF-ß) and IL-10 (p < 0.001 and p < 0.05, respectively), key cytokines used by Treg cells to exert their immunosuppressive functions. Our findings suggest the contribution of both TGF-ß and IL-10 in the suppressive activity of Treg cells.


Asunto(s)
Infecciones por VIH , VIH-1 , Monocitos , Linfocitos T Reguladores , Humanos , Linfocitos T Reguladores/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , VIH-1/inmunología , VIH-1/fisiología , Monocitos/inmunología , Masculino , Polilisina/análogos & derivados , Polilisina/farmacología , Adulto , Poli I-C/inmunología , Poli I-C/farmacología , Femenino , Persona de Mediana Edad , Carboximetilcelulosa de Sodio/análogos & derivados , Factor de Crecimiento Transformador beta/metabolismo , Interleucina-10/metabolismo , Activación de Linfocitos/inmunología , Citocinas/metabolismo , Interleucina-6/metabolismo , Inmunocompetencia , Factor de Necrosis Tumoral alfa/metabolismo , Células Cultivadas
8.
Gut Microbes ; 16(1): 2412669, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39397690

RESUMEN

Gut microbiota-derived metabolites play a pivotal role in the maintenance of intestinal immune homeostasis. Here, we demonstrate that the human commensal Clostridium sporogenes possesses a specific metabolic fingerprint, consisting predominantly of the tryptophan catabolite indole-3-propionic acid (IPA), the branched-chain acids (BCFAs) isobutyrate and isovalerate and the short-chain fatty acids (SCFAs) acetate and propionate. Mono-colonization of germ-free mice with C. sporogenes (CS mice) affected colonic mucosal immune cell phenotypes, including up-regulation of Il22 gene expression, and increased abundance of transcriptionally active colonic tuft cells and Foxp3+ regulatory T cells (Tregs). In DSS-induced colitis, conventional mice suffered severe inflammation accompanied by loss of colonic crypts. These symptoms were absent in CS mice. In conventional, but not CS mice, bulk RNAseq analysis of the colon revealed an increase in inflammatory and Th17-related gene signatures. C. sporogenes-derived IPA reduced IL-17A protein expression by suppressing mTOR activity and by altering ribosome-related pathways in Th17 cells. Additionally, BCFAs and SCFAs generated by C. sporogenes enhanced the activity of Tregs and increased the production of IL-22, which led to protection from colitis. Collectively, we identified C. sporogenes as a therapeutically relevant probiotic bacterium that might be employed in patients with inflammatory bowel disease (IBD).


Asunto(s)
Clostridium , Colitis , Colon , Microbioma Gastrointestinal , Interleucina-22 , Linfocitos T Reguladores , Células Th17 , Animales , Ratones , Clostridium/metabolismo , Colitis/microbiología , Colitis/inducido químicamente , Colitis/inmunología , Colitis/metabolismo , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Colon/microbiología , Colon/inmunología , Colon/metabolismo , Ratones Endogámicos C57BL , Interleucinas/metabolismo , Interleucinas/genética , Humanos , Sulfato de Dextran , Interleucina-17/metabolismo , Ácidos Grasos Volátiles/metabolismo
9.
Arch Dermatol Res ; 316(9): 655, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39352524

RESUMEN

Pemphigus is an IgG-mediated autoimmune condition characterized by autoantibodies targeting desmogleins, leading to acantholysis. Current treatments, including systemic corticosteroids and immunosuppressive drugs, are associated with significant adverse effects. Mesenchymal stem cells (MSCs) offer a promising alternative due to their immunomodulatory properties and low immunogenicity. This study evaluates the immunomodulatory effects of dental follicle mesenchymal stem cells (DF-MSCs) obtained from healthy donors on Pemphigus vulgaris (PV) patients and healthy controls by examining T-cell proliferation, apoptosis, cytokine levels, and anti-desmoglein 1/3 IgG profiles. Peripheral blood mononuclear cells (PBMCs) were isolated from twenty-one symptomatic PV patients and eleven healthy volunteers. DF-MSCs were characterized and differentiated into osteocytes, adipocytes, and chondrocytes. Peripheral blood mononuclear cells (PBMCs) were co-cultured with DF-MSCs, and various assays were conducted to evaluate T-cell proliferation, apoptosis, regulatory T cells, cytokine expression, and autoantibody levels. Results showed that DF-MSC co-cultures significantly reduced lymphocyte proliferation (43.58-16.27%), IL-4 (38.06 ng/L to 32.26 ng/L), TNF-α (32.45 ng/L to 29.41 ng/L), and DSG1 (3.29 ng/ml to 3.00 ng/ml) and DSG3 (262.40 ng/ml to 245.08 ng/ml) levels in PV patients. An increase in regulatory T cells (1.22-3.75%), IL-10 (47.46 pg/ml to 54.94 pg/ml), and IFN-γ (12.39 ng/ml to 19.70 ng/ml) was also observed. No significant changes were noted in healthy controls. These findings suggest that DF-MSCs could potentially offer a curative approach for treating pemphigus by restoring immune balance. However, further clinical trials are necessary to confirm their efficacy.


Asunto(s)
Autoanticuerpos , Proliferación Celular , Células Madre Mesenquimatosas , Pénfigo , Humanos , Pénfigo/inmunología , Pénfigo/terapia , Pénfigo/sangre , Células Madre Mesenquimatosas/inmunología , Femenino , Masculino , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Adulto , Técnicas de Cocultivo , Desmogleína 3/inmunología , Apoptosis/inmunología , Saco Dental/inmunología , Células Cultivadas , Citocinas/metabolismo , Persona de Mediana Edad , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Diferenciación Celular/inmunología , Desmogleína 1/inmunología , Leucocitos Mononucleares/inmunología , Linfocitos T Reguladores/inmunología , Estudios de Casos y Controles
10.
Cell Mol Life Sci ; 81(1): 423, 2024 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-39367914

RESUMEN

Active vitamin D, known for its role in promoting osteoporosis, has immunomodulatory effects according to the latest evidence. Eldecalcitol (ED-71) is a representative of the third-generation novel active vitamin D analogs, and its specific immunological mechanisms in ameliorating diabetic osteoporosis remain unclear. We herein evaluated the therapeutic effects of ED-71 in the context of type 2 diabetes mellitus (T2DM), delving into its underlying mechanisms. In a T2DM mouse model, ED-71 attenuated bone loss and marrow adiposity. Simultaneously, it rectified imbalanced glucose homeostasis and dyslipidemia, ameliorated pancreatic ß-cell damage and hepatic glycolipid metabolism disorder. Subsequently, in mice injected with the Treg cell-depleting agent CD25, we observed that the beneficial effects of ED-71 mentioned earlier were partially contingent on the Treg subsets ratio. Mechanistically, ED-71 promoted the differentiation of CD4+ T cells into Treg subsets, facilitating Ca2+ influx and the expression of ORAI1 and STIM1, pivotal proteins in store-operated Ca2+ entry (SOCE). The SOCE inhibitor, 2-APB, partially attenuated the positive effects of ED-71 observed in the above results. Overall, ED-71 regulates SOCE-mediated Treg cell differentiation, accomplishing the dual purpose of simultaneously ameliorating diabetic osteoporosis and glucolipid metabolic disorders, showcasing its potential in osteoimmunity therapy and interventions for diseases involving SOCE.


Asunto(s)
Diferenciación Celular , Diabetes Mellitus Tipo 2 , Osteoporosis , Linfocitos T Reguladores , Vitamina D , Animales , Masculino , Ratones , Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/complicaciones , Glucolípidos/farmacología , Glucolípidos/uso terapéutico , Ratones Endogámicos C57BL , Proteína ORAI1/metabolismo , Osteoporosis/tratamiento farmacológico , Osteoporosis/metabolismo , Osteoporosis/patología , Molécula de Interacción Estromal 1/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/inmunología , Vitamina D/análogos & derivados , Vitamina D/farmacología , Vitamina D/uso terapéutico
11.
Front Immunol ; 15: 1393418, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39364408

RESUMEN

Osteoarthritis (OA) is a prevalent clinical condition affecting the entire joint, characterized by its multifactorial etiology and complex pathophysiology. The onset of OA is linked to inflammatory mediators produced by the synovium, cartilage, and subchondral bone, all of which are closely tied to cartilage degradation. Consequently, OA may also be viewed as a systemic inflammatory disorder. Emerging studies have underscored the significance of T cells in the development of OA. Notably, imbalances in Th1/Th2 and Th17/Treg immune cells may play a crucial role in the pathogenesis of OA. This review aims to compile recent advancements in understanding the role of T cells and their Th/Treg subsets in OA, examines the immune alterations and contributions of Th/Treg cells to OA progression, and proposes novel directions for future research, including potential therapeutic strategies for OA.


Asunto(s)
Osteoartritis , Linfocitos T Reguladores , Humanos , Osteoartritis/inmunología , Osteoartritis/etiología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
12.
Oncoimmunology ; 13(1): 2412874, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39376580

RESUMEN

Recent findings revealed that neoantigen-specific cytotoxic type 1 regulatory T (TR1) CD4 T cells can subvert cancer immunotherapy by killing type 1 conventional dendritic cells (cDC1s) that present tumor antigens bound to MHC class II. This underlines the importance of cDC1s for eliciting anticancer immunity but poses a novel clinical challenge.


Asunto(s)
Células Dendríticas , Inmunoterapia , Neoplasias , Linfocitos T Reguladores , Humanos , Células Dendríticas/inmunología , Linfocitos T Reguladores/inmunología , Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Animales , Antígenos de Neoplasias/inmunología
13.
BMC Musculoskelet Disord ; 25(1): 791, 2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-39375626

RESUMEN

BACKGROUND: Postmenopausal osteoporosis (PMO) results from a reduction in bone mass and microarchitectural deterioration in bone tissue due to estrogen deficiency, which may increase the incidence of fragility fractures. In recent years, the "gut-immune response-bone" axis has been proposed as a novel potential approach in the prevention and treatment of PMO. Studies on ovariectomized murine model indicated the reciprocal role of Th17 cells and Treg cells in the aetiology of osteoporosis. However, the relationship among gut microbiota, immune cells and bone metabolic indexes remains unknown in PMO. METHODS: A total of 77 postmenopausal women were recruited for the study and divided into control (n = 30), osteopenia (n = 19), and osteoporosis (n = 28) groups based on their T score. The frequency of Treg and Th17 cells in lymphocytes were analyzed by flow cytometry. The serum levels of interleukin (IL)-10, 17 A, 1ß, 6, tumor necrosis factor (TNF)-α, and lipopolysaccharide (LPS) were determined via enzyme-linked immunosorbent assay. Additionally, 16S rRNA gene V3-V4 region sequencing analysis was performed to investigate the gut microbiota of the participants. RESULTS: The results demonstrated decreased bacterial richness and diversed intestinal composition in PMO. In addition, significant differences of relative abundance of the gut microbial community in phylum and genus levels were found, mainly including increased Bacteroidota, Proteobacteria, and Campylobacterota, as well as reduced Firmicutes, Butyricicoccus, and Faecalibacterium. Intriugingly, in the osteoporosis group, the concentration of Treg cells and associated IL-10 in peripheral circulation was negatively regulated, while other chronic systemic proinflammatory cytokines and Th17 cells showed opposite trends. Moreover, significantly elevated plasma lipopolysaccharide (LPS) in patients with osteoporosis indicated that disrupted intestinal integrity and permeability. A correlation analysis showed close relationships between gut bacteria and inflammation. CONCLUSIONS: Collectively, these observations will lead to a better understanding of the relationship among bone homeostasis, the microbiota, and circulating immune cells in PMO. The elevated LPS levels of osteoporosis patients which not only indicate a breach in intestinal integrity but also suggest a novel biomarker for assessing osteoporosis risk linked to gut health.


Asunto(s)
Microbioma Gastrointestinal , Osteoporosis Posmenopáusica , Linfocitos T Reguladores , Células Th17 , Humanos , Femenino , Microbioma Gastrointestinal/inmunología , Persona de Mediana Edad , Osteoporosis Posmenopáusica/inmunología , Osteoporosis Posmenopáusica/microbiología , Osteoporosis Posmenopáusica/sangre , Anciano , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Movimiento Celular , Citocinas/sangre , Posmenopausia/inmunología
14.
Front Immunol ; 15: 1440388, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39380998

RESUMEN

Introduction: Failure to adequate decidualization leads to adverse pregnancy outcomes including pregnancy loss. Although there are plenty of reports underscoring immune dysfunction as the main cause of abortion in CBA/J females mated with DBA/2 males (CBA/J × DBA/2), little is known about the potential role of impaired endometrial decidualization. Methods: Endometrial stromal cells (ESCs) from CBA/J mice were in-vitro decidualized, and the proteome profile of the secretome was investigated by membrane-based array. CBA/J mice were perfused In-utero with either decidualized ESCs (C×D/D), undecidualized ESCs (C×D/ND), or PBS (C×D/P) 12 days before mating with DBA/2 males. Control mice were not manipulated and were mated with male DBA/2 (C×D) or Balb/c (C×B) mice. On day 13.5 of pregnancy, reproductive parameters were measured. In-vivo tracking of EdU-labeled ESCs was performed using fluorescence microscopy. The frequency of regulatory T cells (Tregs) in paraaortic/renal and inguinal lymph nodes was measured by flow cytometry. The proliferation of pregnant CBA/J splenocytes in response to stimulation with DBA/2 splenocytes was assessed by 5,6-carboxyfluorescein diacetate succinimidyl ester (CFSE) flow cytometry. Results: In C×D/D mice, the resorption rate was reduced to match that seen in the C×B group. Intrauterine perfused ESCs appeared in uterine stroma after 2 days, which remained there for at least 12 days. There was no difference in the number of implantation sites and embryo weight across all groups. The frequency of Tregs in the inguinal lymph nodes was similar across all groups, but it increased in the paraaortic/renal lymph nodes of C×D/D mice to the level found in C×B mice. No significant changes were observed in the proliferation of splenocytes from pregnant C×D/D compared to those of the C×D group in response to stimulation with DBA/2 splenocytes. Decidualization of ESCs was associated with a profound alteration in ESC secretome exemplified by alteration in proteins involved in extracellular matrix (ECM) remodeling, response to inflammation, senescence, and immune cell trafficking. Discussion: Our results showed that the deficiency of Tregs is not the primary driver of abortion in the CBA/J × DBA/2 model and provided evidence that impaired endometrial decidualization probably triggers endometrial immune dysfunction and abortion in this model.


Asunto(s)
Aborto Espontáneo , Decidua , Endometrio , Ratones Endogámicos CBA , Ratones Endogámicos DBA , Células del Estroma , Linfocitos T Reguladores , Femenino , Animales , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Embarazo , Ratones , Células del Estroma/metabolismo , Células del Estroma/inmunología , Endometrio/inmunología , Endometrio/metabolismo , Masculino , Decidua/inmunología , Decidua/metabolismo , Aborto Espontáneo/inmunología , Modelos Animales de Enfermedad , Ratones Endogámicos BALB C
15.
Front Immunol ; 15: 1463971, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39351219

RESUMEN

Introduction: In this study, we report a novel therapeutic approach redirecting antigen-specific CD4+ T cells recognizing a hybrid insulin peptide (BDC2.5 T cell receptor (TCR) transgenic CD4+ T cells) to attract and suppress islet-specific CD8+ T cells T cells in the non-obese diabetic (NOD) mouse model, and prevent the development of autoimmune diabetes. Methods: Purified BDC2.5 CD4+ T cells were induced to differentiate into regulatory T cells (Tregs). The Tregs were then electroporated with mRNA encoding chimeric human ß2 microglobulin (hß2m) covalently linked to insulin B chain amino acids 15-23 (designated INS-eTreg) or islet-specific glucose-6-phosphatase related protein (IGRP) peptide 206-214 (designated IGRP-eTreg). Immunoregulatory functions of these engineered regulatory T cells (eTregs) were tested by in vitro assays and in vivo co-transfer experiments with ß-cell-antigen-specific CD8+ T cells in NOD.Scid mice or by adoptive transfer into young, pre-diabetic NOD mice. Results: These eTregs were phenotyped by flow cytometry, and shown to have high expression of FoxP3, as well as other markers of Treg function, including IL-10. They suppressed polyclonal CD4+ T cells and antigen-specific CD8+ T cells (recognizing insulin or IGRP), decreasing proliferation and increasing exhaustion and regulatory markers in vitro. In vivo, eTregs reduced diabetes development in co-transfer experiments with pathogenic antigen-specific CD8+ T cells (INS-CD8+ or IGRP-CD8+ cells) into NOD.Scid mice. Finally, when the eTreg were injected into young NOD mice, they reduced insulitis and prevented spontaneous diabetes in the recipient mice. Conclusion: Our results suggest a novel therapeutic strategy to protect NOD mice by targeting antigen-specific cytotoxic CD8+ T cells, using redirected antigen-specific CD4+ Treg cells, to suppress autoimmune diabetes. This may suggest an innovative therapy for protection of people at risk of development of type 1 diabetes.


Asunto(s)
Linfocitos T CD8-positivos , Diabetes Mellitus Tipo 1 , Ratones Endogámicos NOD , Linfocitos T Reguladores , Animales , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Linfocitos T CD8-positivos/inmunología , Linfocitos T Reguladores/inmunología , Ratones , Humanos , Femenino , Ratones SCID , Insulina/inmunología , Traslado Adoptivo , Ratones Transgénicos , Glucosa-6-Fosfatasa/inmunología , Glucosa-6-Fosfatasa/genética , Microglobulina beta-2/genética , Microglobulina beta-2/inmunología
16.
Front Cell Infect Microbiol ; 14: 1437207, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39386167

RESUMEN

Introduction: Pulmonary tuberculosis (PTB) remains one of the deadliest infectious diseases. Understanding PTB immunity is of potential value for exploring immunotherapy for treating chemotherapy-resistant PTB. CD4+CD25+Foxp3+ regulatory T cells (Tregs) are key players that impair immune responses to Mycobacteria tuberculosis (MTB). Currently, the intrinsic factors governing Treg expansion and influencing the immunosuppressive attributes of Tregs in PTB patients are far from clear. Methods: Here, we employed flow cytometry to determine the frequency of Tregs and the expression of B and T lymphocyte attenuator (BTLA) and its ligand, herpesvirus entry mediator (HVEM), on Tregs in patients with active PTB. Furthermore, the expression of conventional T cells and of programmed death-ligand 1 (PD-L1) and programmed death-1 (PD-1) on Tregs in patients with active PTB was determined. We then examined the characteristics of BTLA/HVEM expression and its correlation with Treg frequency and PD-L1 and PD-1 expression on Tregs in PTB patients. Results: The frequency of Tregs was increased in PTB patients and it had a relevance to PTB progression. Intriguingly, the axis of cosignal molecules, BTLA and HVEM, were both downregulated on the Tregs of PTB patients compared with healthy controls (HCs), which was the opposite of their upregulation on conventional T cells. Unexpectedly, their expression levels were positively correlated with the frequency of Tregs, respectively. These seemingly contradictory results may be interpreted as follows: the downregulation of BTLA and HVEM may alleviate BTLA/HVEM cis-interaction-mediated coinhibitory signals pressing on naïve Tregs, helping their activation, while the BTLA/HVEM axis on effector Tregs induces a costimulatory signal, promoting their expansion. Certainly, the mechanism underlying such complex effects remains to be explored. Additionally, PD-L1 and PD-1, regarded as two of the markers characterizing the immunosuppressive attributes and differentiation potential of Tregs, were upregulated on the Tregs of PTB patients. Further analysis revealed that the expression levels of BTLA and HVEM were positively correlated with the frequency of PD-1+Tregs and PD-L1+Tregs, respectively. Conclusion: Our study illuminated distinct characteristics of BTLA/HVEM axis expression on Tregs and uncovered its impact on the expansion and attributes of Tregs in patients with active PTB. Therefore, blockade of the BTLA/HVEM axis may be a promising potential pathway to reduce Treg expansion for the improvement of anti-MTB immune responses.


Asunto(s)
Antígeno B7-H1 , Receptor de Muerte Celular Programada 1 , Receptores Inmunológicos , Miembro 14 de Receptores del Factor de Necrosis Tumoral , Linfocitos T Reguladores , Tuberculosis Pulmonar , Humanos , Linfocitos T Reguladores/inmunología , Receptores Inmunológicos/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Tuberculosis Pulmonar/inmunología , Masculino , Femenino , Adulto , Receptor de Muerte Celular Programada 1/metabolismo , Persona de Mediana Edad , Antígeno B7-H1/metabolismo , Mycobacterium tuberculosis/inmunología , Adulto Joven , Citometría de Flujo
17.
Front Immunol ; 15: 1466901, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39386201

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is the most common and severe form of pulmonary fibrosis, characterized by scar formation in the lung interstitium. Transforming growth factor beta (TGF-ß) is known as a key mediator in the fibrotic process, acting on fibroblasts and mediating their proliferation and differentiation into myofibroblasts. Although the immune system is not considered responsible for the initiation of IPF, markers of tolerogenic immunity define the pro-fibrotic microenvironment in the lungs. In homeostatic conditions, regulatory T cells (Tregs) constitute the main lymphoid population responsible for maintaining peripheral tolerance. Similar to Tregs, regulatory B cells (Bregs) represent a recently described subset of B lymphocytes with immunosuppressive functions. In the context of IPF, numerous studies have suggested a role for Tregs in enhancing fibrosis, mainly via the secretion of TGF-ß. In humans, most studies show increased percentages of Tregs associated with the severity of IPF, although their exact role remains unclear. In mice, the most commonly used model involves triggering acute lung inflammation with bleomycin, leading to a subsequent fibrotic process. Consequently, data are still conflicting, as Tregs may play a protective role during the inflammatory phase and a deleterious role during the fibrotic phase. Bregs have been less studied in the context of IPF, but their role appears to be protective in experimental models of lung fibrosis. This review presents the latest updates on studies exploring the implication of regulatory lymphoid cells in IPF and compares the different approaches to better understand the origins of conflicting findings.


Asunto(s)
Linfocitos B Reguladores , Fibrosis Pulmonar Idiopática , Linfocitos T Reguladores , Fibrosis Pulmonar Idiopática/inmunología , Fibrosis Pulmonar Idiopática/patología , Fibrosis Pulmonar Idiopática/metabolismo , Humanos , Linfocitos T Reguladores/inmunología , Animales , Linfocitos B Reguladores/inmunología , Linfocitos B Reguladores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Pulmón/inmunología , Pulmón/patología , Ratones , Modelos Animales de Enfermedad
18.
Front Endocrinol (Lausanne) ; 15: 1425235, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39391872

RESUMEN

Introduction: The incidence of type-1 diabetes is on the rise, particularly in developed nations, and predominantly affects the youth. While genetic predisposition plays a substantial role, environmental factors, including alterations in the gut microbiota, are increasingly recognized as significant contributors to the disease. Methods: In this study, we utilized germ-free non-obese diabetic mice to explore the effects of microbiota colonization during early life on type-1 diabetes susceptibility. Results: Our findings reveal that microbiota introduction at birth, rather than at weaning, significantly reduces the risk of type-1 diabetes, indicating a crucial window for microbiota-mediated modulation of immune responses. This protective effect was independent of alterations in intestinal barrier function but correlated with testosterone levels in male mice. Additionally, early life colonization modulated T cell subset frequencies, particularly T helper cells and regulatory T cells, in the intestine, potentially shaping type-1 diabetes predisposition. Discussion: Our findings underscore the pivotal role of early-life microbial interactions in immune regulation and the development of autoimmune diseases.


Asunto(s)
Diabetes Mellitus Tipo 1 , Microbioma Gastrointestinal , Ratones Endogámicos NOD , Animales , Diabetes Mellitus Tipo 1/microbiología , Diabetes Mellitus Tipo 1/inmunología , Ratones , Masculino , Femenino , Linfocitos T Reguladores/inmunología
19.
Methods Cell Biol ; 189: 169-188, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39393882

RESUMEN

Multiparametric flow cytometry (MFC) represents an essential tool for immune monitoring, and validation of MFC panels is a fundamental prerequisite in routine laboratory settings as well as for translational and clinical research purposes. Regulatory T cells (TREGs) constitute a subset of CD4+ effector T cells that modulate the immune response in numerous settings, including autoimmune disease, allergy, microbial infection, tumor immunity, transplantation, and more. These cells comprise a small fraction of total CD4+ T cells in human peripheral blood and mouse spleen. In oncology, TREG cells are highly relevant, as they are involved in the suppression of the anti-tumor response in many types of cancer, to the extent that the first immune checkpoint inhibitor approved for clinical use in humans was a monoclonal antibody directed against CTLA-4, a molecule functionally associated with TREGs. Due to all these factors, robust assays are mandatory to accurately determine TREG cell frequency and function. Here, we describe the validation of an 8-color flow-cytometry protocol for TREG detection and analysis in a real-world laboratory scenario. The entire process includes the workflow plan and the standard operating procedure resembling each phase, from the panel design to the staining, acquisition, and analysis steps. Validation is planned to be performed in replicates on fresh whole blood samples derived from multiple healthy subjects. The analytical validity of the TREG cell assay is ensured by testing the intra-assay accuracy. The detailed procedure for the entire process is accompanied by important troubleshooting suggestions and other useful tips.


Asunto(s)
Citometría de Flujo , Linfocitos T Reguladores , Citometría de Flujo/métodos , Citometría de Flujo/normas , Linfocitos T Reguladores/inmunología , Humanos , Animales , Ratones
20.
JCI Insight ; 9(19)2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39377224

RESUMEN

T follicular regulatory (TFR) cells are found in the germinal center (GC) response and, along with T follicular helper (TFH) cells, help to control the development of high-affinity antibodies (Ab). Although TFR cells are generally thought to repress GC B cells and the Ab response, we have previously shown that in a mouse food allergy model, TFR cells produce IL-10 and play an essential helper role such that in the absence of TFR cells, IgE responses are diminished. Here we show that in this food allergy response, TFR cells produced IL-4 that promotes the generation of antigen-specific IgE. We show that food allergy-primed TFR cells specifically upregulate IL-4 gene transcription and produce functional IL-4 that promoted IgE responses both in vitro and in vivo. We determined that IgE responses are dependent on a high level of IL-4 produced by follicular T cells in the GC, explaining the need for IL-4 produced by TFR cells in the food allergy response. Overall, our findings have demonstrated that in food allergy, TFR cells can produce IL-4 and regulate IgE in a manner that augments the role of TFH cells in IgE responses.


Asunto(s)
Hipersensibilidad a los Alimentos , Centro Germinal , Inmunoglobulina E , Interleucina-4 , Linfocitos T Reguladores , Animales , Interleucina-4/inmunología , Interleucina-4/metabolismo , Inmunoglobulina E/inmunología , Hipersensibilidad a los Alimentos/inmunología , Ratones , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Centro Germinal/inmunología , Modelos Animales de Enfermedad , Linfocitos B/inmunología , Células T Auxiliares Foliculares/inmunología , Femenino , Interleucina-10/metabolismo , Interleucina-10/inmunología , Ratones Noqueados , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA