Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38.436
Filtrar
1.
Nat Commun ; 15(1): 3884, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38719909

RESUMEN

Only a minority of cancer patients benefit from immune checkpoint blockade therapy. Sophisticated cross-talk among different immune checkpoint pathways as well as interaction pattern of immune checkpoint molecules carried on circulating small extracellular vesicles (sEV) might contribute to the low response rate. Here we demonstrate that PD-1 and CD80 carried on immunocyte-derived sEVs (I-sEV) induce an adaptive redistribution of PD-L1 in tumour cells. The resulting decreased cell membrane PD-L1 expression and increased sEV PD-L1 secretion into the circulation contribute to systemic immunosuppression. PD-1/CD80+ I-sEVs also induce downregulation of adhesion- and antigen presentation-related molecules on tumour cells and impaired immune cell infiltration, thereby converting tumours to an immunologically cold phenotype. Moreover, synchronous analysis of multiple checkpoint molecules, including PD-1, CD80 and PD-L1, on circulating sEVs distinguishes clinical responders from those patients who poorly respond to anti-PD-1 treatment. Altogether, our study shows that sEVs carry multiple inhibitory immune checkpoints proteins, which form a potentially targetable adaptive loop to suppress antitumour immunity.


Asunto(s)
Antígeno B7-1 , Antígeno B7-H1 , Vesículas Extracelulares , Receptor de Muerte Celular Programada 1 , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Humanos , Antígeno B7-1/metabolismo , Antígeno B7-H1/metabolismo , Antígeno B7-H1/inmunología , Animales , Ratones , Línea Celular Tumoral , Femenino , Neoplasias/inmunología , Neoplasias/patología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Tolerancia Inmunológica , Ratones Endogámicos C57BL , Masculino , Microambiente Tumoral/inmunología
2.
Sci Immunol ; 9(95): eadi4191, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38728412

RESUMEN

Conventional dendritic cells (DCs) are essential mediators of antitumor immunity. As a result, cancers have developed poorly understood mechanisms to render DCs dysfunctional within the tumor microenvironment (TME). After identification of CD63 as a specific surface marker, we demonstrate that mature regulatory DCs (mregDCs) migrate to tumor-draining lymph node tissues and suppress DC antigen cross-presentation in trans while promoting T helper 2 and regulatory T cell differentiation. Transcriptional and metabolic studies showed that mregDC functionality is dependent on the mevalonate biosynthetic pathway and its master transcription factor, SREBP2. We found that melanoma-derived lactate activates SREBP2 in tumor DCs and drives conventional DC transformation into mregDCs via homeostatic or tolerogenic maturation. DC-specific genetic silencing and pharmacologic inhibition of SREBP2 promoted antitumor CD8+ T cell activation and suppressed melanoma progression. CD63+ mregDCs were found to reside within the lymph nodes of several preclinical tumor models and in the sentinel lymph nodes of patients with melanoma. Collectively, this work suggests that a tumor lactate-stimulated SREBP2-dependent program promotes CD63+ mregDC development and function while serving as a promising therapeutic target for overcoming immune tolerance in the TME.


Asunto(s)
Células Dendríticas , Ácido Láctico , Ratones Endogámicos C57BL , Transducción de Señal , Proteína 2 de Unión a Elementos Reguladores de Esteroles , Células Dendríticas/inmunología , Animales , Ratones , Humanos , Proteína 2 de Unión a Elementos Reguladores de Esteroles/inmunología , Ácido Láctico/metabolismo , Transducción de Señal/inmunología , Melanoma/inmunología , Melanoma/patología , Progresión de la Enfermedad , Tolerancia Inmunológica/inmunología , Femenino , Línea Celular Tumoral , Microambiente Tumoral/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/patología
3.
Front Immunol ; 15: 1336599, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38715621

RESUMEN

Introduction: Pompe disease, a lysosomal storage disorder, is characterized by acid α-glucosidase (GAA) deficiency and categorized into two main subtypes: infantile-onset Pompe disease (IOPD) and late-onset Pompe disease (LOPD). The primary treatment, enzyme replacement therapy (ERT) with recombinant human GAA (rhGAA), faces challenges due to immunogenic responses, including the production of anti-drug antibody (ADA), which can diminish therapeutic efficacy. This study aims to assess the effectiveness of immune tolerance induction (ITI) therapy in cross-reactive immunologic material (CRIM)-positive Pompe disease patients with established high ADA levels. Method: In a single-center, open-label prospective study, we assessed ITI therapy's efficacy in Pompe disease patients, both IOPD and LOPD, with persistently elevated ADA titers (≥1:12,800) and clinical decline. The ITI regimen comprised bortezomib, rituximab, methotrexate, and intravenous immunoglobulin. Biochemical data, biomarkers, ADA titers, immune status, and respiratory and motor function were monitored over six months before and after ITI. Results: This study enrolled eight patients (5 IOPD and 3 LOPD). After a 6-month ITI course, median ADA titers significantly decreased from 1:12,800 (range 1:12,800-1:51,200) to 1:1,600 (range 1:400-1:12,800), with sustained immune tolerance persisting up to 4.5 years in some cases. Serum CK levels were mostly stable or decreased, stable urinary glucose tetrasaccharide levels were maintained in four patients, and no notable deterioration in respiratory or ambulatory status was noted. Adverse events included two treatable infection episodes and transient symptoms like numbness and diarrhea. Conclusion: ITI therapy effectively reduces ADA levels in CRIM-positive Pompe disease patients with established high ADA titers, underscoring the importance of ADA monitoring and timely ITI initiation. The findings advocate for personalized immunogenicity risk assessments to enhance clinical outcomes. In some cases, prolonged immune suppression may be necessary, highlighting the need for further studies to optimize ITI strategies for Pompe disease treatment. ClinicalTrials.gov NCT02525172; https://clinicaltrials.gov/study/NCT02525172.


Asunto(s)
Terapia de Reemplazo Enzimático , Enfermedad del Almacenamiento de Glucógeno Tipo II , Tolerancia Inmunológica , alfa-Glucosidasas , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , alfa-Glucosidasas/uso terapéutico , alfa-Glucosidasas/inmunología , alfa-Glucosidasas/administración & dosificación , Terapia de Reemplazo Enzimático/efectos adversos , Terapia de Reemplazo Enzimático/métodos , Enfermedad del Almacenamiento de Glucógeno Tipo II/inmunología , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Inmunoglobulinas Intravenosas/uso terapéutico , Inmunoglobulinas Intravenosas/administración & dosificación , Metotrexato/uso terapéutico , Metotrexato/administración & dosificación , Estudios Prospectivos , Rituximab/uso terapéutico , Rituximab/efectos adversos , Rituximab/administración & dosificación , Resultado del Tratamiento
4.
Front Immunol ; 15: 1393283, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38742111

RESUMEN

For decades, innate immune cells were considered unsophisticated first responders, lacking the adaptive memory of their T and B cell counterparts. However, mounting evidence demonstrates the surprising complexity of innate immunity. Beyond quickly deploying specialized cells and initiating inflammation, two fascinating phenomena - endotoxin tolerance (ET) and trained immunity (TI) - have emerged. ET, characterized by reduced inflammatory response upon repeated exposure, protects against excessive inflammation. Conversely, TI leads to an enhanced response after initial priming, allowing the innate system to mount stronger defences against subsequent challenges. Although seemingly distinct, these phenomena may share underlying mechanisms and functional implications, blurring the lines between them. This review will delve into ET and TI, dissecting their similarities, differences, and the remaining questions that warrant further investigation.


Asunto(s)
Endotoxinas , Tolerancia Inmunológica , Inmunidad Innata , Memoria Inmunológica , Humanos , Animales , Endotoxinas/inmunología , Inflamación/inmunología , Inmunidad Adaptativa , Inmunidad Entrenada
5.
Oncoimmunology ; 13(1): 2352179, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38746869

RESUMEN

Cancer-associated fibroblasts (CAFs) exhibit remarkable phenotypic heterogeneity, with specific subsets implicated in immunosuppression in various malignancies. However, whether and how they attenuate anti-tumor immunity in gastric cancer (GC) remains elusive. CPT1C, a unique isoform of carnitine palmitoyltransferase pivotal in regulating fatty acid oxidation, is briefly indicated as a protumoral metabolic mediator in the tumor microenvironment (TME) of GC. In the present study, we initially identified specific subsets of fibroblasts exclusively overexpressing CPT1C, hereby termed them as CPT1C+CAFs. Subsequent findings indicated that CPT1C+CAFs fostered a stroma-enriched and immunosuppressive TME as they correlated with extracellular matrix-related molecular features and enrichment of both immunosuppressive subsets, especially M2-like macrophages, and multiple immune-related pathways. Next, we identified that CPT1C+CAFs promoted the M2-like phenotype of macrophage in vitro. Bioinformatic analyses unveiled the robust IL-6 signaling between CPT1C+CAFs and M2-like phenotype of macrophage and identified CPT1C+CAFs as the primary source of IL-6. Meanwhile, suppressing CPT1C expression in CAFs significantly decreased IL-6 secretion in vitro. Lastly, we demonstrated the association of CPT1C+CAFs with therapeutic resistance. Notably, GC patients with high CPT1C+CAFs infiltration responded poorly to immunotherapy in clinical cohort. Collectively, our data not only present the novel identification of CPT1C+CAFs as immunosuppressive subsets in TME of GC, but also reveal the underlying mechanism that CPT1C+CAFs impair tumor immunity by secreting IL-6 to induce the immunosuppressive M2-like phenotype of macrophage in GC.


Asunto(s)
Fibroblastos Asociados al Cáncer , Carnitina O-Palmitoiltransferasa , Interleucina-6 , Macrófagos , Microambiente Tumoral , Humanos , Microambiente Tumoral/inmunología , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/inmunología , Fibroblastos Asociados al Cáncer/patología , Interleucina-6/metabolismo , Interleucina-6/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/patología , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Fenotipo , Animales , Ratones , Masculino , Femenino , Línea Celular Tumoral , Tolerancia Inmunológica
6.
Am J Reprod Immunol ; 91(5): e13859, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38722063

RESUMEN

Recurrent Spontaneous Abortion (RSA) is a common pregnancy complication, that has multifactorial causes, and currently, 40%-50% of cases remain unexplained, referred to as Unexplained RSA (URSA). Due to the elusive etiology and mechanisms, clinical management is exceedingly challenging. In recent years, with the progress in reproductive immunology, a growing body of evidence suggests a relationship between URSA and maternal-fetal immunology, offering hope for the development of tailored treatment strategies. This article provides an immunological perspective on the pathogenesis, diagnosis, and treatment of RSA. On one hand, it comprehensively reviews the immunological mechanisms underlying RSA, including abnormalities in maternal-fetal interface immune tolerance, maternal-fetal interface immune cell function, gut microbiota-mediated immune dysregulation, and vaginal microbiota-mediated immune anomalies. On the other hand, it presents the diagnosis and existing treatment modalities for RSA. This article offers a clear knowledge framework for understanding RSA from an immunological standpoint. In conclusion, while the "layers of the veil" regarding immunological factors in RSA are gradually being unveiled, our current research may only scratch the surface. In terms of immunological etiology, effective diagnostic tools for RSA are currently lacking, and the efficacy and safety of immunotherapies, primarily based on lymphocyte immunotherapy and intravenous immunoglobulin, remain contentious.


Asunto(s)
Aborto Habitual , Humanos , Femenino , Embarazo , Aborto Habitual/inmunología , Tolerancia Inmunológica , Intercambio Materno-Fetal/inmunología , Microbioma Gastrointestinal/inmunología , Inmunoterapia/métodos
7.
Ann Allergy Asthma Immunol ; 132(5): 592-593, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38702104
8.
Cancer Rep (Hoboken) ; 7(5): e2066, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38703051

RESUMEN

BACKGROUND: The tumor microenvironment of solid tumors governs the differentiation of otherwise non-immunosuppressive macrophages and gamma delta (γδ) T cells into strong immunosuppressors while promoting suppressive abilities of known immunosuppressors such as myeloid-derived suppressor cells (MDSCs) upon infiltration into the tumor beds. RECENT FINDINGS: In epithelial malignancies, tumor-associated macrophages (TAMs), precursor monocytic MDSCs (M-MDSCs), and gamma delta (γδ) T cells often acquire strong immunosuppressive abilities that dampen spontaneous immune responses by tumor-infiltrating T cells and B lymphocytes against cancer. Both M-MDSCs and γδ T cells have been associated with worse prognosis for multiple epithelial cancers. CONCLUSION: Here we discuss recent discoveries on how tumor-associated macrophages and precursor M-MDSCs as well as tumor associated-γδ T cells acquire immunosuppressive abilities in the tumor beds, promote cancer metastasis, and perspectives on how possible novel interventions could restore the effective adaptive immune responses in epithelial cancers.


Asunto(s)
Linfocitos Infiltrantes de Tumor , Células Supresoras de Origen Mieloide , Microambiente Tumoral , Humanos , Microambiente Tumoral/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Células Supresoras de Origen Mieloide/inmunología , Linfocitos Intraepiteliales/inmunología , Neoplasias Glandulares y Epiteliales/inmunología , Neoplasias Glandulares y Epiteliales/patología , Tolerancia Inmunológica , Animales , Macrófagos Asociados a Tumores/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Células Mieloides/inmunología
9.
Proc Natl Acad Sci U S A ; 121(20): e2320268121, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38709934

RESUMEN

Insulin is a central autoantigen in the pathogenesis of T1D, and thymic epithelial cell expression of insulin under the control of the Autoimmune Regulator (Aire) is thought to be a key component of maintaining tolerance to insulin. In spite of this general working model, direct detection of this thymic selection on insulin-specific T cells has been somewhat elusive. Here, we used a combination of highly sensitive T cell receptor transgenic models for detecting thymic selection and sorting and sequencing of Insulin-specific CD4+ T cells from Aire-deficient mice as a strategy to further define their selection. This analysis revealed a number of unique t cell receptor (TCR) clones in Aire-deficient hosts with high affinity for insulin/major histocompatibility complex (MHC) ligands. We then modeled the thymic selection of one of these clones in Aire-deficient versus wild-type hosts and found that this model clone could escape thymic negative selection in the absence of thymic Aire. Together, these results suggest that thymic expression of insulin plays a key role in trimming and removing high-affinity insulin-specific T cells from the repertoire to help promote tolerance.


Asunto(s)
Proteína AIRE , Insulina , Receptores de Antígenos de Linfocitos T , Timo , Factores de Transcripción , Animales , Timo/inmunología , Timo/metabolismo , Timo/citología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Ratones , Insulina/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Tolerancia Inmunológica , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Ratones Transgénicos , Ratones Noqueados , Células Clonales , Ratones Endogámicos C57BL
10.
Int J Mol Sci ; 25(9)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38732104

RESUMEN

Hypertensive disorders of pregnancy (HDP), including preeclampsia (PE) and gestational hypertension (GH), are major causes of maternal and foetal morbidity and mortality. This review elucidates the role of regulatory T cells (Tregs) in the immunological aspects of HDP and explores their therapeutic potential. Tregs, which play a critical role in maintaining immune homeostasis, are crucial in pregnancy to prevent immune-mediated rejection of the foetus. The review highlights that Tregs contribute to immunological adaptation in normal pregnancy, ensuring foetal acceptance. In contrast, HDP is associated with Treg dysfunction, which is marked by decreased numbers and impaired regulatory capacity, leading to inadequate immune tolerance and abnormal placental development. This dysfunction is particularly evident in PE, in which Tregs fail to adequately modulate the maternal immune response against foetal antigens, contributing to the pathophysiology of the disorder. Therapeutic interventions aiming to modulate Treg activity represent a promising avenue for HDP management. Studies in animal models and limited clinical trials suggest that enhancing Treg functionality could mitigate HDP symptoms and improve pregnancy outcomes. However, given the multifactorial nature of HDP and the intricate regulatory mechanisms of Tregs, the review explores the complexities of translating in vitro and animal model findings into effective clinical therapies. In conclusion, while the precise role of Tregs in HDP is still being unravelled, their central role in immune regulation during pregnancy is indisputable. Further research is needed to fully understand the mechanisms by which Tregs contribute to HDP and to develop targeted therapies that can safely and effectively harness their regulatory potential for treating hypertensive diseases of pregnancy.


Asunto(s)
Hipertensión Inducida en el Embarazo , Linfocitos T Reguladores , Humanos , Linfocitos T Reguladores/inmunología , Embarazo , Femenino , Hipertensión Inducida en el Embarazo/inmunología , Hipertensión Inducida en el Embarazo/terapia , Animales , Preeclampsia/inmunología , Preeclampsia/terapia , Tolerancia Inmunológica
11.
Int Immunopharmacol ; 133: 112130, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38648712

RESUMEN

Neutrophils and T lymphocytes are closely related to occurrence of immunosuppression in sepsis. Studies have shown that neutrophil apoptosis decreases and T lymphocyte apoptosis increases in sepsis immunosuppression, but the specific mechanism involved remains unclear. In the present study, we found Toll-like Receptor 2 (TLR2) and programmed death-ligand 1 (PD-L1) were significantly activated in bone marrow neutrophils of wild-type mice after LPS treatment and that they were attenuated by treatment with C29, an inhibitor of TLR2. PD-L1 activation inhibits neutrophil apoptosis, whereas programmed death protein 1 (PD-1)activation promotes apoptosis of T lymphocytes, which leads to immunosuppression. Mechanistically, when sepsis occurs, pro-inflammatory factors and High mobility group box-1 protein (HMGB1) passively released from dead cells cause the up-regulation of PD-L1 through TLR2 on neutrophils. The binding of PD-L1 and PD-1 on T lymphocytes leads to increased apoptosis of T lymphocytes and immune dysfunction, eventually resulting in the occurrence of sepsis immunosuppression. In vivo experiments showed that the HMGB1 inhibitor glycyrrhizic acid (GA) and the TLR2 inhibitor C29 could inhibit the HMGB1/TLR2/PD-L1 pathway, and improving sepsis-induced lung injury. In summary, this study shows that HMGB1 regulates PD-L1 and PD-1 signaling pathways through TLR2, which leads to immunosuppression.


Asunto(s)
Apoptosis , Antígeno B7-H1 , Proteína HMGB1 , Ratones Endogámicos C57BL , Neutrófilos , Sepsis , Linfocitos T , Receptor Toll-Like 2 , Animales , Receptor Toll-Like 2/metabolismo , Proteína HMGB1/metabolismo , Sepsis/inmunología , Sepsis/metabolismo , Antígeno B7-H1/metabolismo , Apoptosis/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/efectos de los fármacos , Ratones , Masculino , Tolerancia Inmunológica , Ácido Glicirrínico/farmacología , Ácido Glicirrínico/uso terapéutico , Lipopolisacáridos/inmunología , Transducción de Señal , Terapia de Inmunosupresión
12.
J Neuroimmunol ; 390: 578347, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38663308

RESUMEN

Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system resulting from loss of immune tolerance. Many disease-modifying therapies for MS have broad immunosuppressive effects on peripheral immune cells, but this can increase risks of infection and attenuate vaccine-elicited immunity. A more targeted approach is to re-establish immune tolerance in an autoantigen-specific manner. This review discusses methods to achieve this, focusing on tolerogenic dendritic cells. Clinical trials in other autoimmune diseases also provide learnings with regards to clinical translation of this approach, including identification of autoantigen(s), selection of appropriate patients and administration route and frequency.


Asunto(s)
Células Dendríticas , Tolerancia Inmunológica , Inmunoterapia , Esclerosis Múltiple , Humanos , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/terapia , Animales , Tolerancia Inmunológica/inmunología , Inmunoterapia/métodos , Autoantígenos/inmunología
13.
Int Immunopharmacol ; 133: 112166, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38678673

RESUMEN

Dendritic cells (DCs) are specialized antigen-presenting cells that play an important role in inducing and maintaining immune tolerance. The altered distribution and/or function of DCs contributes to defective tolerance in autoimmune diseases such as type 1 diabetes (T1D). In human T1D and in NOD mouse models, DCs share some defects and are often described as less tolerogenic and excessively immunogenic. In the NOD mouse model, the autoimmune response is associated with a defect in the Stat5b signaling pathway. We have reported that expressing a constitutively active form of Stat5b in DCs of transgenic NOD mice (NOD.Stat5b-CA), re-established their tolerogenic function, restored autoimmune tolerance and conferred protection from diabetes. However, the role and molecular mechanisms of Stat5b signaling in regulating splenic conventional DCs tolerogenic signature remained unclear. In this study, we reported that, compared to immunogenic splenic DCs of NOD, splenic DCs of NOD.Stat5b-CA mice exhibited a tolerogenic profile marked by elevated PD-L1 and PD-L2 expression, reduced pro-inflammatory cytokine production, increased frequency of the cDC2 subset and decreased frequency of the cDC1 subset. This tolerogenic profile was associated with increased Ezh2 and IRF4 but decreased IRF8 expression. We also found an upregulation of PD-L1 in the cDC1 subset and high PD-L1 and PD-L2 expression in cDC2 of NOD.Stat5b-CA mice. Mechanistically, we demonstrated that Ezh2 plays an important role in the maintenance of high PD-L1 expression in cDC1 and cDC2 subsets and that Ezh2 inhibition resulted in PD-L1 but not PD-L2 downregulation which was more drastic in the cDC2 subset. Additionally, Ezh2 inhibition severely reduced the cDC2 subset and increased the cDC1 subset and Stat5b-CA.DC pro-inflammatory cytokine production. Together our data suggest that the Stat5b-Ezh2 axis is critical for the maintenance of tolerogenic high PD-L1-expressing cDC2 and autoimmune tolerance in NOD.Stat5b-CA mice.


Asunto(s)
Antígeno B7-H1 , Células Dendríticas , Diabetes Mellitus Tipo 1 , Proteína Potenciadora del Homólogo Zeste 2 , Tolerancia Inmunológica , Ratones Endogámicos NOD , Factor de Transcripción STAT5 , Animales , Células Dendríticas/inmunología , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Factor de Transcripción STAT5/metabolismo , Factor de Transcripción STAT5/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Diabetes Mellitus Tipo 1/inmunología , Ratones , Humanos , Transducción de Señal , Femenino , Ratones Transgénicos , Citocinas/metabolismo , Células Cultivadas
14.
Trials ; 25(1): 272, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38641837

RESUMEN

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.


Asunto(s)
Pollos , Hipersensibilidad a los Alimentos , Niño , Lactante , Bovinos , Humanos , Femenino , Animales , Hipersensibilidad a los Alimentos/diagnóstico , Hipersensibilidad a los Alimentos/prevención & control , Leche/efectos adversos , Alérgenos/efectos adversos , Tolerancia Inmunológica
15.
Front Immunol ; 15: 1359914, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38646539

RESUMEN

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.


Asunto(s)
Neoplasias Gastrointestinales , Transducción de Señal , Humanos , Neoplasias Gastrointestinales/inmunología , Neoplasias Gastrointestinales/patología , Neoplasias Gastrointestinales/metabolismo , Animales , Metástasis de la Neoplasia , Tolerancia Inmunológica , Escape del Tumor
16.
Immun Inflamm Dis ; 12(4): e1246, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38668747

RESUMEN

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.


Asunto(s)
Tolerancia Inmunológica , Inmunoglobulina E , Hipersensibilidad a la Leche , Proteínas de la Leche , Pruebas Cutáneas , Humanos , Hipersensibilidad a la Leche/inmunología , Hipersensibilidad a la Leche/epidemiología , Hipersensibilidad a la Leche/diagnóstico , Hipersensibilidad a la Leche/sangre , Masculino , Lactante , Femenino , Perú/epidemiología , Proteínas de la Leche/inmunología , Inmunoglobulina E/inmunología , Inmunoglobulina E/sangre , Animales , Alérgenos/inmunología , Bovinos , Heces , Complejo de Antígeno L1 de Leucocito/análisis
17.
Thromb Res ; 237: 196-202, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38640641

RESUMEN

INTRODUCTION: The most notable challenge facing hemophilia A treatment is the development of inhibitors against factor VIII, resulting in increased clinical and socioeconomic burdens due to the need for expensive bypassing agents (BPAs). Although immune tolerance induction (ITI) is currently the primary approach for inhibiting and reducing the inhibitors, the lengthy duration of ITI necessitates the continued use of BPA to manage bleeding episodes. In this study, we aimed to obtain real-world evidence on the clinical and economic aspects and associated burdens experienced by patients with hemophilia A with inhibitors undergoing ITI in Korea. METHODS: Claims data from January 1, 2007, to December 31, 2020, were used in this study. The study cohort comprised patients with hemophilia A undergoing ITI, who were categorized into three groups: successful, failed, or continuation of ITI. We evaluated clinical and economic burdens, including monthly healthcare visits, medication costs, and total medical expenses. RESULTS: The study involved 33 cases of ITI across 32 patients. Excluding seven continuation cases where success could not be determined at the observation point, the estimated success rate of ITI was 80.8 %. The median duration of ITI for all patients was 25.7 months. While no significant disparities were noted in the ITI duration between successful and unsuccessful cases (24.51 vs. 25.66 months), substantial discrepancies were observed in the duration of BPA usage (11.10 vs. 25.66 months) and the number of prescribed BPAs (1.79 vs. 2.97). CONCLUSION: Successful ITI reduced both clinical and economic burdens, resulting in decreased monthly medication expenses and overall medical costs.


Asunto(s)
Hemofilia A , Tolerancia Inmunológica , Humanos , Hemofilia A/economía , Hemofilia A/inmunología , Hemofilia A/tratamiento farmacológico , República de Corea , Masculino , Niño , Adulto , Adolescente , Preescolar , Factor VIII/uso terapéutico , Factor VIII/inmunología , Factor VIII/economía , Costo de Enfermedad , Adulto Joven , Femenino , Lactante , Costos de la Atención en Salud
18.
Mol Biol Rep ; 51(1): 571, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38662155

RESUMEN

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.


Asunto(s)
Transformación Celular Neoplásica , Galectinas , Receptor 2 Celular del Virus de la Hepatitis A , Leucemia , Humanos , Galectinas/metabolismo , Leucemia/inmunología , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/genética , Animales , Tolerancia Inmunológica , Transducción de Señal , Escape del Tumor , Proliferación Celular , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo
19.
Cancer Immunol Res ; 12(4): 393-399, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38562083

RESUMEN

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.


Asunto(s)
Neoplasias , Linfocitos T Reguladores , Humanos , Neoplasias/terapia , Tolerancia Inmunológica , Inmunoterapia , Inflamación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA