Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.053
Filtrar
Más filtros











Intervalo de año de publicación
1.
Front Immunol ; 15: 1362120, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38962016

RESUMEN

Cancer stem cells (CSCs), accounting for only a minor cell proportion (< 1%) within tumors, have profound implications in tumor initiation, metastasis, recurrence, and treatment resistance due to their inherent ability of self-renewal, multi-lineage differentiation, and tumor-initiating potential. In recent years, accumulating studies indicate that CSCs and tumor immune microenvironment act reciprocally in driving tumor progression and diminishing the efficacy of cancer therapies. Extracellular vesicles (EVs), pivotal mediators of intercellular communications, build indispensable biological connections between CSCs and immune cells. By transferring bioactive molecules, including proteins, nucleic acids, and lipids, EVs can exert mutual influence on both CSCs and immune cells. This interaction plays a significant role in reshaping the tumor immune microenvironment, creating conditions favorable for the sustenance and propagation of CSCs. Deciphering the intricate interplay between CSCs and immune cells would provide valuable insights into the mechanisms of CSCs being more susceptible to immune escape. This review will highlight the EV-mediated communications between CSCs and each immune cell lineage in the tumor microenvironment and explore potential therapeutic opportunities.


Asunto(s)
Vesículas Extracelulares , Neoplasias , Células Madre Neoplásicas , Microambiente Tumoral , Microambiente Tumoral/inmunología , Humanos , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/metabolismo , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Animales , Comunicación Celular/inmunología , Escape del Tumor , Inmunomodulación
2.
J Clin Immunol ; 44(7): 156, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38954150

RESUMEN

BACKGROUND: Thymoma presents with several autoimmune manifestations and is associated with secondary autoimmune regulator (AIRE) deficiency. Pneumonitis has recently been described as an autoimmune manifestation associated with thymoma presenting with similar clinical, radiographic, histological, and autoantibody features as seen in patients with inherited AIRE deficiency who suffer from Autoimmune PolyEndocrinopathy-Candidiasis-Ectodermal Dystrophy (APECED) syndrome. OBJECTIVES: To treat two patients with biopsy-proven thymoma-associated pneumonitis with lymphocyte-directed immunomodulation. METHODS: Two patients with thymoma were enrolled on IRB-approved protocols at the NIH Clinical Center. We performed history and physical examination; laboratory, radiographic, histologic and pulmonary function evaluations; and measurement of the lung-directed autoantibodies KCNRG and BPIFB1 prior to and at 1- and 6-months following initiation of lymphocyte-directed immunomodulation with azathioprine with or without rituximab. RESULTS: Combination T- and B-lymphocyte-directed immunomodulation resulted in improvement of clinical, functional, and radiographic parameters at 6-month follow-up evaluations in both patients with sustained remission up to 12-36 months following treatment initiation. CONCLUSION: Lymphocyte-directed immunomodulation remitted autoimmune pneumonitis in two patients with thymoma.


Asunto(s)
Inmunomodulación , Timoma , Humanos , Timoma/inmunología , Timoma/complicaciones , Timoma/diagnóstico , Femenino , Masculino , Rituximab/uso terapéutico , Autoanticuerpos/inmunología , Persona de Mediana Edad , Neoplasias del Timo/inmunología , Neoplasias del Timo/complicaciones , Neoplasias del Timo/diagnóstico , Neumonía/etiología , Neumonía/inmunología , Neumonía/diagnóstico , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/etiología , Adulto , Azatioprina/uso terapéutico , Linfocitos B/inmunología , Resultado del Tratamiento , Linfocitos T/inmunología
3.
Theranostics ; 14(9): 3739-3759, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38948054

RESUMEN

Background: The repair of osteoporotic bone defects remains challenging due to excessive reactive oxygen species (ROS), persistent inflammation, and an imbalance between osteogenesis and osteoclastogenesis. Methods: Here, an injectable H2-releasing hydrogel (magnesium@polyethylene glycol-poly(lactic-co-glycolic acid), Mg@PEG-PLGA) was developed to remodel the challenging bone environment and accelerate the repair of osteoporotic bone defects. Results: This Mg@PEG-PLGA gel shows excellent injectability, shape adaptability, and phase-transition ability, can fill irregular bone defect areas via minimally invasive injection, and can transform into a porous scaffold in situ to provide mechanical support. With the appropriate release of H2 and magnesium ions, the 2Mg@PEG-PLGA gel (loaded with 2 mg of Mg) displayed significant immunomodulatory effects through reducing intracellular ROS, guiding macrophage polarization toward the M2 phenotype, and inhibiting the IκB/NF-κB signaling pathway. Moreover, in vitro experiments showed that the 2Mg@PEG-PLGA gel inhibited osteoclastogenesis while promoting osteogenesis. Most notably, in animal experiments, the 2Mg@PEG-PLGA gel significantly promoted the repair of osteoporotic bone defects in vivo by scavenging ROS and inhibiting inflammation and osteoclastogenesis. Conclusions: Overall, our study provides critical insight into the design and development of H2-releasing magnesium-based hydrogels as potential implants for repairing osteoporotic bone defects.


Asunto(s)
Regeneración Ósea , Hidrogeles , Hidrógeno , Magnesio , Osteogénesis , Osteoporosis , Polietilenglicoles , Especies Reactivas de Oxígeno , Animales , Magnesio/química , Magnesio/administración & dosificación , Especies Reactivas de Oxígeno/metabolismo , Ratones , Polietilenglicoles/química , Hidrogeles/química , Osteoporosis/tratamiento farmacológico , Osteogénesis/efectos de los fármacos , Hidrógeno/farmacología , Hidrógeno/administración & dosificación , Hidrógeno/química , Células RAW 264.7 , Regeneración Ósea/efectos de los fármacos , Inmunomodulación/efectos de los fármacos , Andamios del Tejido/química , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Poliésteres
4.
J Colloid Interface Sci ; 672: 179-199, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-38838627

RESUMEN

Mesenchymal stem cell-derived exosomes (MSC-Exos) have emerged as promising candidates for cell-free therapy in tissue regeneration. However, the native osteogenic and angiogenic capacities of MSC-Exos are often insufficient to repair critical-sized bone defects, and the underlying immune mechanisms remain elusive. Furthermore, achieving sustained delivery and stable activity of MSC-Exos at the defect site is essential for optimal therapeutic outcomes. Here, we extracted exosomes from osteogenically pre-differentiated human bone marrow mesenchymal stem cells (hBMSCs) by ultracentrifugation and encapsulated them in gelatin methacryloyl (GelMA) hydrogel to construct a composite scaffold. The resulting exosome-encapsulated hydrogel exhibited excellent mechanical properties and biocompatibility, facilitating sustained delivery of MSC-Exos. Osteogenic pre-differentiation significantly enhanced the osteogenic and angiogenic properties of MSC-Exos, promoting osteogenic differentiation of hBMSCs and angiogenesis of human umbilical vein endothelial cells (HUVECs). Furthermore, MSC-Exos induced polarization of Raw264.7 cells from a pro-inflammatory phenotype to an anti-inflammatory phenotype under simulated inflammatory conditions, thereby creating an immune microenvironment conducive to osteogenesis. RNA sequencing and bioinformatics analysis revealed that MSC-Exos activate the p53 pathway through targeted delivery of internal microRNAs and regulate macrophage polarization by reducing DNA oxidative damage. Our study highlights the potential of osteogenic exosome-encapsulated composite hydrogels for the development of cell-free scaffolds in bone tissue engineering.


Asunto(s)
Regeneración Ósea , Diferenciación Celular , Exosomas , Gelatina , Hidrogeles , Inmunomodulación , Células Madre Mesenquimatosas , Osteogénesis , Exosomas/química , Exosomas/metabolismo , Células Madre Mesenquimatosas/citología , Gelatina/química , Osteogénesis/efectos de los fármacos , Hidrogeles/química , Hidrogeles/farmacología , Regeneración Ósea/efectos de los fármacos , Humanos , Ratones , Diferenciación Celular/efectos de los fármacos , Animales , Inmunomodulación/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Células RAW 264.7 , Metacrilatos/química , Metacrilatos/farmacología , Tamaño de la Partícula , Células Cultivadas , Propiedades de Superficie , Neovascularización Fisiológica/efectos de los fármacos , Andamios del Tejido/química
5.
J Cell Mol Med ; 28(11): e18410, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38853457

RESUMEN

Troponin T1 (TNNT1) plays a crucial role in muscle contraction but its role in cancer, particularly in kidney renal clear cell carcinoma (KIRC), is not well-understood. This study explores the expression, clinical significance and biological functions of TNNT1 in various cancers, with an emphasis on its involvement in KIRC. We analysed TNNT1 expression in cancers using databases like TCGA and GTEx, assessing its prognostic value, mutation patterns, methylation status and functional implications. The study also examined TNNT1's effect on the tumour microenvironment and drug sensitivity in KIRC, complemented by in vitro TNNT1 knockdown experiments in KIRC cells. TNNT1 is overexpressed in several cancers and linked to adverse outcomes, showing frequent upregulation mutations and abnormal methylation. Functionally, TNNT1 connects to muscle and cancer pathways, affects immune infiltration and drug responses, and its overexpression in KIRC is associated with advanced disease and reduced survival. Knocking down TNNT1 curbed KIRC cell growth. TNNT1's aberrant expression plays a significant role in tumorigenesis and immune modulation, highlighting its value as a prognostic biomarker and a potential therapeutic target in KIRC and other cancers. Further studies are essential to understand TNNT1's oncogenic mechanisms in KIRC.


Asunto(s)
Carcinogénesis , Carcinoma de Células Renales , Regulación Neoplásica de la Expresión Génica , Neoplasias Renales , Troponina T , Humanos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinogénesis/genética , Carcinogénesis/inmunología , Carcinogénesis/patología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/metabolismo , Línea Celular Tumoral , Proliferación Celular , Metilación de ADN , Inmunomodulación/genética , Neoplasias Renales/genética , Neoplasias Renales/inmunología , Neoplasias Renales/patología , Neoplasias Renales/metabolismo , Mutación/genética , Pronóstico , Troponina T/metabolismo , Troponina T/genética , Microambiente Tumoral/inmunología
6.
Front Immunol ; 15: 1396827, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38855102

RESUMEN

Glucocorticoids, which have long served as fundamental therapeutics for diverse inflammatory conditions, are still widely used, despite associated side effects limiting their long-term use. Among their key mediators is glucocorticoid-induced leucine zipper (GILZ), recognized for its anti-inflammatory and immunosuppressive properties. Here, we explore the immunomodulatory effects of GILZ in macrophages through transcriptomic analysis and functional assays. Bulk RNA sequencing of GILZ knockout and GILZ-overexpressing macrophages revealed significant alterations in gene expression profiles, particularly impacting pathways associated with the inflammatory response, phagocytosis, cell death, mitochondrial function, and extracellular structure organization activity. GILZ-overexpression enhances phagocytic and antibacterial activity against Salmonella typhimurium and Escherichia coli, potentially mediated by increased nitric oxide production. In addition, GILZ protects macrophages from pyroptotic cell death, as indicated by a reduced production of reactive oxygen species (ROS) in GILZ transgenic macrophages. In contrast, GILZ KO macrophages produced more ROS, suggesting a regulatory role of GILZ in ROS-dependent pathways. Additionally, GILZ overexpression leads to decreased mitochondrial respiration and heightened matrix metalloproteinase activity, suggesting its involvement in tissue remodeling processes. These findings underscore the multifaceted role of GILZ in modulating macrophage functions and its potential as a therapeutic target for inflammatory disorders, offering insights into the development of novel therapeutic strategies aimed at optimizing the benefits of glucocorticoid therapy while minimizing adverse effects.


Asunto(s)
Macrófagos , Mitocondrias , Fagocitosis , Piroptosis , Factores de Transcripción , Animales , Mitocondrias/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Inmunomodulación , Especies Reactivas de Oxígeno/metabolismo , Ratones Noqueados , Glucocorticoides/farmacología , Ratones Endogámicos C57BL , Salmonella typhimurium/inmunología , Escherichia coli/inmunología
7.
Clin Transl Med ; 14(6): e1735, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38899748

RESUMEN

BACKGROUND: Mitochondrial outer membrane permeabilisation (MOMP) plays a pivotal role in cellular death and immune activation. A deeper understanding of the impact of tumour MOMP on immunity will aid in guiding more effective immunotherapeutic strategies. METHODS: A comprehensive pan-cancer dataset comprising 30 cancer-type transcriptomic cohorts, 20 immunotherapy transcriptomic cohorts and three immunotherapy scRNA-seq datasets was collected and analysed to determine the influence of tumour MOMP activity on clinical prognosis, immune infiltration and immunotherapy effectiveness. Leveraging 65 scRNA-Seq datasets, the MOMP signature (MOMP.Sig) was developed to accurately reflect tumour MOMP activity. The clinical predictive value of MOMP.Sig was explored through machine learning models. Integration of the MOMP.Sig model and a pan-cancer immunotherapy CRISPR screen further investigated potential targets to overcome immunotherapy resistance, which subsequently underwent clinical validation. RESULTS: Our research revealed that elevated MOMP activity reduces mortality risk in cancer patients, drives the formation of an anti-tumour immune environment and enhances the response to immunotherapy. This finding emphasises the potential clinical application value of MOMP activity in immunotherapy. MOMP.Sig, offering a more precise indicator of tumour cell MOMP activity, demonstrated outstanding predictive efficacy in machine-learning models. Moreover, with the assistance of the MOMP.Sig model, FOXO1 was identified as a core modulator that promotes immune resistance. Finally, these findings were successfully validated in clinical immunotherapy cohorts of skin cutaneous melanoma and triple-negative breast cancer patients. CONCLUSIONS: This study enhances our understanding of MOMP activity in immune modulation, providing valuable insights for more effective immunotherapeutic strategies across diverse tumours.


Asunto(s)
Inmunoterapia , Membranas Mitocondriales , Neoplasias , Humanos , Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Membranas Mitocondriales/metabolismo , Inmunomodulación/efectos de los fármacos
8.
Sci Rep ; 14(1): 14092, 2024 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890401

RESUMEN

Oral squamous cell carcinoma (OSCC) is one of the most common malignant tumours, warranting novel treatments. Here, we examined the therapeutic efficacy of inhibiting p21 activated kinase 4 (PAK4) in OSCC and determined its immunomodulatory effect by focusing on the enhancement of anti-tumour effects. We examined PAK4 expression in OSCC cells and human clinical samples and analysed the proliferation and apoptosis of OSCC cells following PAK4 inhibition in vitro. We also investigated the effects of in vivo administration of a PAK4 inhibitor on immune cell distribution and T-cell immune responses in OSCC tumour-bearing mice. PAK4 was detected in all OSCC cells and OSCC tissue samples. PAK4 inhibitor reduced the proliferation of OSCC cells and induced apoptosis. PAK4 inhibitor significantly attenuated tumour growth in mouse and was associated with increased proportions of IFN-γ-producing CD8+ T-cells. Furthermore, PAK4 inhibitor increased the number of dendritic cells (DCs) and up-regulated the surface expression of various lymphocyte co-stimulatory molecules, including MHC-class I molecules, CD80, CD83, CD86, and CD40. These DCs augmented CD8+ T-cell activation upon co-culture. Our results suggest that PAK4 inhibition in OSCC can have direct anti-tumour and immunomodulatory effects, which might benefit the treatment of this malignancy.


Asunto(s)
Carcinoma de Células Escamosas , Proliferación Celular , Inmunomodulación , Neoplasias de la Boca , Quinasas p21 Activadas , Quinasas p21 Activadas/metabolismo , Quinasas p21 Activadas/antagonistas & inhibidores , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/inmunología , Neoplasias de la Boca/patología , Neoplasias de la Boca/metabolismo , Humanos , Animales , Ratones , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , Inmunomodulación/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Línea Celular Tumoral , Apoptosis/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Femenino , Masculino
9.
Immun Inflamm Dis ; 12(6): e1311, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38874280

RESUMEN

BACKGROUND: The heterogeneity of tumor endothelial cells (TECs) hinders the efficacy of antiangiogenic therapies (AATs). Only a small percentage of angiogenic TECs are considered effective targets for AATs. Immunomodulatory ECs (IMECs), as a newly focused functional subgroup of endothelial cells (ECs), are being evaluated for their ability to regulate tumor immune balance and influence existing AATs. METHODS: Based on single-cell transcriptome data from colorectal cancer in a publicly available database, we conducted a wide array of bioinformatic approaches to study EC subsets that meet the IMECs definition. Our investigation encompassed the gene expression signatures of these subsets, cellular composition differences, cell-cell interactions. RESULTS: Two subsets that meet the IMECs definition were found in tumors and para-cancerous tissues. Combined with the results of gene ontological analysis and interaction with CD4+ T cells, we found that IMECs can present MHC-II antigens to mature CD4+ T cells. There were differences in the level of interaction between IMECs and different types of mature CD4+ T cell subsets. In addition, IMEC subsets had different expression levels of angiogenesis related genes. The angiogenesis score of IMECs decreased after patients received immunotherapy. IMEC subsets do not depend on a single proangiogenic receptor and are involved in regulating angiogenesis, which may reduce the efficacy of AATs. The adverse effects of specific IMEC subsets on AATs were validated in the RNA-seq dataset of the bevacizumab treatment group. CONCLUSION: Our study suggests the potential MHC-II antigen presentation capacity of IMECs and the enhanced angiogenesis characteristics within tumors. The function of IMECs in the vascular network may have a potentially adverse effect on AATs. Controlling the functional properties of IMECs may be a new angle for tumor therapy.


Asunto(s)
Neoplasias Colorrectales , Células Endoteliales , Análisis de la Célula Individual , Transcriptoma , Humanos , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/terapia , Células Endoteliales/metabolismo , Células Endoteliales/inmunología , Presentación de Antígeno , Neovascularización Patológica/inmunología , Neovascularización Patológica/genética , Resistencia a Antineoplásicos , Perfilación de la Expresión Génica , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Inmunomodulación , Linfocitos T CD4-Positivos/inmunología
10.
Cell Biochem Funct ; 42(5): e4081, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38934382

RESUMEN

Butyrophilin (BTN) proteins are a type of membrane protein that belongs to the Ig superfamily. They exhibit a high degree of structural similarity to molecules in the B7 family. They fulfill a complex function in regulating immune responses, including immunomodulatory roles, as they influence γδ T cells. The biology of BTN molecules indicates that they are capable of inhibiting the immune system's ability to detect antigens within tumors. A dynamic association between BTN molecules and cellular surfaces is also recognized in specific contexts, influencing their biology. Notably, the dynamism of BTN3A1 is associated with the immunosuppression of T cells or the activation of Vγ9Vδ2 T cells. Cancer immunotherapy relies heavily on T cells to modulate immune function within the intricate interaction of the tumor microenvironment (TME). A significant interaction between the TME and antitumor immunity involves the presence of BTN, which should be taken into account when developing immunotherapy. This review explores potential therapeutic applications of BTN molecules, based on the current understanding of their biology.


Asunto(s)
Butirofilinas , Inmunomodulación , Neoplasias , Humanos , Butirofilinas/metabolismo , Butirofilinas/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Inmunomodulación/efectos de los fármacos , Inmunoterapia , Microambiente Tumoral/inmunología
11.
Life Sci ; 351: 122786, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38848944

RESUMEN

Gangliosides represent glycolipids containing sialic acid residues, present on the cell membrane with glycan residues exposed to the extracellular matrix (ECM), while the ceramides are anchored within the membrane. These molecules play a critical role in pathophysiological processes such as host-pathogen interactions, cell-cell recognition, signal transduction, cell adhesion, motility, and immunomodulation. Accumulated evidence suggests the overexpression of gangliosides on tumor tissues in comparison to healthy human tissues. These tumor-associated gangliosides have been implicated in various facets of tumor biology, including cell motility, differentiation, signaling, immunosuppression, angiogenesis, and metastasis. Consequently, these entities emerge as attractive targets for immunotherapeutic interventions. Notably, the administration of antibodies targeting gangliosides has demonstrated cytotoxic effects on cancer cells that exhibit an overexpression of these glycolipids. Passive immunotherapy approaches utilizing murine or murine/human chimeric anti-ganglioside antibodies have been explored as potential treatments for diverse cancer types. Additionally, vaccination strategies employing tumor-associated gangliosides in conjunction with adjuvants have entered the realm of promising techniques currently undergoing clinical trials. The present comprehensive review encapsulates the multifaceted roles of gangliosides in tumor initiation, progression, immunosuppression, and metastasis. Further, an overview is provided of the correlation between the expression status of gangliosides in normal and tumor cells and its impact on cancer patient survival. Furthermore, the discussion extends to ongoing and completed clinical trials employing diverse strategies to target gangliosides, elucidating their effectiveness in treating cancers. This emerging discipline is expected to supply substantial impetus for the establishment of novel therapeutic strategies.


Asunto(s)
Gangliósidos , Inmunomodulación , Inmunoterapia , Neoplasias , Humanos , Gangliósidos/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Animales , Inmunoterapia/métodos
12.
Iran J Immunol ; 21(2): 132-146, 2024 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-38800958

RESUMEN

Background: It is well-known that TH1 and Treg cells exert anti- and pro-tumorigenic activity, respectively. Thus, TH1 cell suppression together with Treg cell hyperactivation contribute to tumor development. Glycyrrhiza glabra (G. glabra) has various immunomodulatory and anti-tumorigenic properties. Objective: To explore the impacts of G. glabra extract on different parameters related to TH1 and Treg cells using a breast cancer (BC) model. Methods: Four groups of Balb/C mice bearing 4T1 cell-induced BC were treated intraperitoneally with either saline or G. glabra extract at dosages of 50, 100 and 150 mg/kg (G. glabra-50, G. glabra-100, and G. glabra-150, respectively). After sacrificing animals on day 26, the frequency of splenic TH1 and Treg cells, the levels of serum IFN-γ, TGF-ß, and IL-12, and intra-tumoral expressions of granzyme-B, T-bet, and FOXP3 were assessed. Results: Compared to untreated tumor control (UTC) group, treatment with G. glabra-50, G. glabra-100, or G. glabra-150 increased the survival rate, percentage of TH1 cells, and T-bet expression. Conversely, they reduced the percentage of Treg cells, and serum TGF-ß levels. In comparison to the UTC group, treatment with G. glabra-50 and G. glabra-150 increased the serum IL-12 levels. Treatment with G. glabra-100 and G. glabra-150 boosted granzyme-B expression. Treatment with G. glabra-150 elevated IFN-γ levels, while treatment with G. glabra-50 decreased the FOXP3 expression. IL-12 levels were higher in mice treated with G. glabra-150 compared to those treated with G. glabra-100. Conclusion: Treatment of mice with BC using G. glabra extract improved survival rate, reduced tumor growth, and modulated T cell-mediated immune responses.


Asunto(s)
Neoplasias de la Mama , Modelos Animales de Enfermedad , Glycyrrhiza , Extractos Vegetales , Linfocitos T Reguladores , Células TH1 , Animales , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Femenino , Ratones , Células TH1/inmunología , Células TH1/efectos de los fármacos , Glycyrrhiza/química , Extractos Vegetales/farmacología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Ratones Endogámicos BALB C , Humanos , Citocinas/metabolismo , Inmunomodulación/efectos de los fármacos
13.
Cancer Med ; 13(10): e7287, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38770637

RESUMEN

Although the development of immunotherapies has been revolutionary in the treatment of several cancers, many cancer types remain unresponsive to immune-based treatment and are largely managed by chemotherapy drugs. However, chemotherapeutics are not infallible and are frequently rendered ineffective as resistance develops from prolonged exposure. Recent investigations have indicated that some chemotherapy drugs have additional functions beyond their normative cytotoxic capacity and are in fact immune-modifying agents. Of the pharmaceuticals with identified immune-editing properties, gemcitabine is well-studied and of interest to clinicians and scientists alike. Gemcitabine is a chemotherapy drug approved for the treatment of multiple cancers, including breast, lung, pancreatic, and ovarian. Because of its broad applications, relatively low toxicity profile, and history as a favorable combinatory partner, there is promise in the recharacterization of gemcitabine in the context of the immune system. Such efforts may allow the identification of suitable immunotherapeutic combinations, wherein gemcitabine can be used as a priming agent to improve immunotherapy efficacy in traditionally insensitive cancers. This review looks to highlight documented immunomodulatory abilities of one of the most well-known chemotherapy agents, gemcitabine, relating to its influence on cells and proteins of the immune system.


Asunto(s)
Antimetabolitos Antineoplásicos , Gemcitabina , Agentes Inmunomoduladores , Neoplasias , Animales , Humanos , Antimetabolitos Antineoplásicos/uso terapéutico , Antimetabolitos Antineoplásicos/farmacología , Gemcitabina/farmacología , Gemcitabina/uso terapéutico , Agentes Inmunomoduladores/uso terapéutico , Agentes Inmunomoduladores/farmacología , Inmunomodulación/efectos de los fármacos , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología
14.
FASEB J ; 38(10): e23644, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38738472

RESUMEN

Tumors typically lack canonical danger signals required to activate adaptive immunity and also frequently employ substantial immunomodulatory mechanisms that downregulate adaptive responses and contribute to escape from immune surveillance. Given the variety of mechanisms involved in shielding tumors from immune recognition, it is not surprising that single-agent immunomodulatory approaches have been largely unsuccessful in generating durable antitumor responses. Here we report a unique combination of immunomodulatory and cytostatic agents that recondition the tumor microenvironment and eliminate complex and/or poor-prognosis tumor types including the non-immunogenic 4T-1 model of TNBC, the aggressive MOC-2 model of HNSCC, and the high-risk MYCN-amplified model of neuroblastoma. A course of therapy optimized for TNBC cured a majority of tumors in both ectopic and orthotopic settings and eliminated metastatic spread in all animals tested at the highest doses. Immune responses were transferable between therapeutic donor and naïve recipient through adoptive transfer, and a sizeable abscopal effect on distant, untreated lesions could be demonstrated experimentally. Similar results were observed in HNSCC and neuroblastoma models, with characteristic remodeling of the tumor microenvironment documented in all model systems. scRNA-seq analysis implicated upregulation of innate immune responses and antigen presentation in tumor cells and the myeloid cell compartment as critical early events. This analysis also highlighted the potential importance of the autonomic nervous system in the governance of inflammatory processes. The data indicate that the targeting of multiple pathways and mechanisms of action can result in substantial synergistic antitumor effects and suggest follow-up in the neoadjuvant setting may be warranted.


Asunto(s)
Microambiente Tumoral , Animales , Ratones , Microambiente Tumoral/inmunología , Línea Celular Tumoral , Neuroblastoma/inmunología , Neuroblastoma/terapia , Neuroblastoma/patología , Femenino , Humanos , Inmunomodulación , Ratones Endogámicos C57BL
15.
Kaohsiung J Med Sci ; 40(6): 520-529, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38712483

RESUMEN

Autoimmune disease is characterized by the proliferation of harmful immune cells, inducing tissue inflammation and ultimately causing organ damage. Current treatments often lack specificity, necessitating high doses, prolonged usage, and high recurrence rates. Therefore, the identification of innovative and safe therapeutic strategies is urgently required. Recent preclinical studies and clinical trials on inflammatory and autoimmune diseases have evidenced the immunosuppressive properties of mesenchymal stromal cells (MSCs). Studies have demonstrated that extracellular vesicles (EV) derived from MSCs can mitigate abnormal autoinflammation while maintaining safety within the diseased microenvironment. This study conducted a systematic review to elucidate the crucial role of MSC-EVs in alleviating autoimmune diseases, particularly focusing on their impact on the underlying mechanisms of autoimmune conditions such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and inflammatory bowel disease (IBD). By specifically examining the regulatory functions of microRNAs (miRNAs) derived from MSC-EVs, the comprehensive study aimed to enhance the understanding related to disease mechanisms and identify potential diagnostic markers and therapeutic targets for these diseases.


Asunto(s)
Enfermedades Autoinmunes , Vesículas Extracelulares , Células Madre Mesenquimatosas , MicroARNs , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/inmunología , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/trasplante , Enfermedades Autoinmunes/terapia , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , MicroARNs/genética , MicroARNs/metabolismo , Lupus Eritematoso Sistémico/terapia , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Artritis Reumatoide/terapia , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Animales , Enfermedades Inflamatorias del Intestino/terapia , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Inmunomodulación
16.
Int Rev Cell Mol Biol ; 386: 167-222, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38782499

RESUMEN

Historically, KRAS has been considered 'undruggable' inspite of being one of the most frequently altered oncogenic proteins in solid tumors, primarily due to the paucity of pharmacologically 'druggable' pockets within the mutant isoforms. However, pioneering developments in drug design capable of targeting the mutant KRAS isoforms especially KRASG12C-mutant cancers, have opened the doors for emergence of combination therapies comprising of a plethora of inhibitors targeting different signaling pathways. SHP2 signaling pathway, primarily known for activation of intracellular signaling pathways such as KRAS has come up as a potential target for such combination therapies as it emerged to be the signaling protein connecting KRAS and the immune signaling pathways and providing the link for understanding the overlapping regions of RAS/ERK/MAPK signaling cascade. Thus, SHP2 inhibitors having potent tumoricidal activity as well as role in immunomodulation have generated keen interest in researchers to explore its potential as combination therapy in KRAS mutant solid tumors. However, the excitement with these combination therapies need to overcome challenges thrown up by drug resistance and enhanced toxicity. In this review, we will discuss KRAS and SHP2 signaling pathways and their roles in immunomodulation and regulation of tumor microenvironment and also analyze the positive effects and drawbacks of the different combination therapies targeted at these signaling pathways along with their present and future potential to treat solid tumors.


Asunto(s)
Inmunomodulación , Neoplasias , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Proto-Oncogénicas p21(ras) , Transducción de Señal , Humanos , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/terapia , Transducción de Señal/efectos de los fármacos , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Inmunomodulación/efectos de los fármacos , Animales , Resultado del Tratamiento , Terapia Molecular Dirigida
17.
J Food Sci ; 89(6): 3829-3846, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38745368

RESUMEN

Lonicera japonica Thunb. (LJT) is known for its valuable medicinal properties that highlight its potential application in the pharmaceutical and health food industry. We predict that LJT polyphenols by network pharmacology may be involved in immunomodulation, and the study of LJT polyphenols regulating immunity is still insufficient; therefore, we experimentally found that LJT enhances immunity by promoting the proliferation and phagocytic activity of RAW246.7 cells. A model of an immunosuppressed mouse was constructed using cyclophosphamide-induced, and LJT was extracted for the intervention. We found that LJT restored immune homeostasis in immune deficiency mice by inhibiting the abnormal apoptosis in lymphocytes, enhancing natural killer cell cytotoxicity, promoting T lymphocyte proliferation, and increasing the CD4+ and CD8+ T lymphocytes in quantity. Moreover, LJT treatment modulates immunity by significantly downregulating lipopolysaccharide-induced inflammation and oxidative stress levels. We verified the immunomodulatory function of LJT through both cell and animal experiments. The combination of potential-protein interactions and molecular docking later revealed that LJT polyphenols were associated with immunomodulatory effects on MAPK1; together, LJT intervention significantly modulates the immune, with the activation of MAPK1 as the underlying mechanism of action, which provided evidence for the utilization of LJT as a nutraceutical in immune function.


Asunto(s)
Inmunomodulación , Lonicera , Farmacología en Red , Extractos Vegetales , Lonicera/química , Animales , Ratones , Extractos Vegetales/farmacología , Farmacología en Red/métodos , Inmunomodulación/efectos de los fármacos , Células RAW 264.7 , Simulación del Acoplamiento Molecular , Polifenoles/farmacología , Proliferación Celular/efectos de los fármacos , Masculino , Apoptosis/efectos de los fármacos , Ratones Endogámicos BALB C
18.
Expert Rev Anti Infect Ther ; 22(6): 435-451, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38804866

RESUMEN

INTRODUCTION: The emergence of antiparasitic drug resistance poses a concerning threat to animals and humans. Mesenchymal Stem Cells (MSCs) have been widely used to treat infections in humans, pets, and livestock. Although this is an emerging field of study, the current review outlines possible mechanisms and examines potential synergism in combination therapies and the possible harmful effects of such an approach. AREAS COVERED: The present study delved into the latest pre-clinical research on utilizing MSCs to treat parasitic infections. As per investigations, the introduction of MSCs to patients grappling with parasitic diseases like schistosomiasis, malaria, cystic echinococcosis, toxoplasmosis, leishmaniasis, and trypanosomiasis has shown a reduction in parasite prevalence. This intervention also alters the levels of both pro- and anti-inflammatory cytokines. Furthermore, the combined administration of MSCs and antiparasitic drugs has demonstrated enhanced efficacy in combating parasites and modulating the immune response. EXPERT OPINION: Mesenchymal stem cells are a potential solution for addressing parasitic drug resistance. This is mainly because of their remarkable immunomodulatory abilities, which can potentially help combat parasites' resistance to drugs.


Asunto(s)
Resistencia a Medicamentos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Enfermedades Parasitarias , Humanos , Animales , Enfermedades Parasitarias/inmunología , Enfermedades Parasitarias/tratamiento farmacológico , Células Madre Mesenquimatosas/inmunología , Antiparasitarios/farmacología , Antiparasitarios/administración & dosificación , Terapia Combinada , Inmunomodulación/efectos de los fármacos , Antiinflamatorios/farmacología , Antiinflamatorios/administración & dosificación , Citocinas/metabolismo , Citocinas/inmunología
19.
Eur J Med Chem ; 274: 116536, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-38805936

RESUMEN

G-quadruplexes (G4s) are commonly formed in the G-rich strand of telomeric DNA. Ligands targeting telomeric G4 induce DNA damage and telomere dysfunction, which makes them potential antitumor drugs. New telomeric G4 ligands with drug-likeness are still needed to be exploited, especially with their antitumor mechanisms thoroughly discussed. In this study, a novel series of quinoxaline analogs were rationally designed and synthesized. Among them, R1 was the most promising ligand for its cytotoxic effects on tumor cells and stabilizing ability with telomeric G4. Cellular assays illustrated that R1 stabilized G4 and induced R-loop accumulation in the telomeric regions, subsequently triggering DNA damage responses, cell cycle arrest in G2/M phase, apoptosis and antiproliferation. Moreover, R1 evoked immunogenic cell death (ICD) in tumor cells, which promoted the maturation of bone marrow derived dendritic cells (BMDCs). In breast cancer mouse model, R1 exhibited a significant decrease in tumor burden through the immunomodulatory effects, including the increase of CD4+ and CD8+ T cells in tumors and cytokine levels in sera. Our research provides a new idea that targeting telomeric G4 induces DNA damage responses, causing antitumor effects both in vitro and in vivo, partially due to the enhancement of immunomodulation.


Asunto(s)
Antineoplásicos , Proliferación Celular , G-Cuádruplex , Quinoxalinas , Telómero , G-Cuádruplex/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Quinoxalinas/química , Quinoxalinas/farmacología , Quinoxalinas/síntesis química , Animales , Humanos , Telómero/efectos de los fármacos , Ligandos , Ratones , Proliferación Celular/efectos de los fármacos , Estructura Molecular , Ensayos de Selección de Medicamentos Antitumorales , Relación Estructura-Actividad , Relación Dosis-Respuesta a Droga , Femenino , Inmunomodulación/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Ratones Endogámicos BALB C , Daño del ADN/efectos de los fármacos
20.
Chem Soc Rev ; 53(11): 5862-5903, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38716589

RESUMEN

Biological nanoparticles, or bionanoparticles, are small molecules manufactured in living systems with complex production and assembly machinery. The products of the assembly systems can be further engineered to generate functionalities for specific purposes. These bionanoparticles have demonstrated advantages such as immune system evasion, minimal toxicity, biocompatibility, and biological clearance. Hence, bionanoparticles are considered the new paradigm in nanoscience research for fabricating safe and effective nanoformulations for therapeutic purposes. Harnessing the power of the immune system to recognize and eradicate malignancies is a viable strategy to achieve better therapeutic outcomes with long-term protection from disease recurrence. However, cancerous tissues have evolved to become invisible to immune recognition and to transform the tumor microenvironment into an immunosuppressive dwelling, thwarting the immune defense systems and creating a hospitable atmosphere for cancer growth and progression. Thus, it is pertinent that efforts in fabricating nanoformulations for immunomodulation are mindful of the tumor-induced immune aberrations that could render cancer nanotherapy inoperable. This review systematically categorizes the immunosuppression mechanisms, the regulatory immunosuppressive cellular players, and critical suppressive molecules currently targeted as breakthrough therapies in the clinic. Finally, this review will summarize the engineering strategies for affording immune moderating functions to bionanoparticles that tip the tumor microenvironment (TME) balance toward cancer elimination, a field still in the nascent stage.


Asunto(s)
Inmunomodulación , Nanopartículas , Neoplasias , Microambiente Tumoral , Humanos , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Neoplasias/inmunología , Microambiente Tumoral/efectos de los fármacos , Inmunomodulación/efectos de los fármacos , Animales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA