Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 152
Filtrar
1.
Sci Rep ; 14(1): 24230, 2024 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-39414902

RESUMEN

Tumor-associated macrophages (TAMs) have been implicated as a tumor microenvironment (TME) cell population, which may be playing a vital role in the inhibition of effective T cell responses in the prostate TME. In this manuscript, we leverage a novel microscale cell culture platform, known as Stacks, to investigate mono-, co-, and tri-culture TME models comprised of prostate tumor cell lines, primary macrophages, and autologous T cells from patients with prostate cancer. Through multiplexed analysis of these multi-cellular prostate tumor models, we capture a dynamic interaction between primary TAMs and activated T cells that resulted in reciprocal proinflammatory activation of both cell populations upon interaction. These findings suggest that activated T cells are capable of reprogramming immunosuppressive TAMs in the context of prostate tumor models and that TAM reprogramming may play a key supportive role in restoring proinflammatory T cell tumor responses in the prostate TME.


Asunto(s)
Movimiento Celular , Reprogramación Celular , Activación de Linfocitos , Neoplasias de la Próstata , Linfocitos T , Microambiente Tumoral , Macrófagos Asociados a Tumores , Masculino , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Humanos , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Reprogramación Celular/inmunología , Activación de Linfocitos/inmunología , Línea Celular Tumoral , Técnicas de Cocultivo , Comunicación Celular/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo
2.
Immunity ; 57(10): 2344-2361.e7, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39321806

RESUMEN

As the most frequent genetic alteration in cancer, more than half of human cancers have p53 mutations that cause transcriptional inactivation. However, how p53 modulates the immune landscape to create a niche for immune escape remains elusive. We found that cancer stem cells (CSCs) established an interleukin-34 (IL-34)-orchestrated niche to promote tumorigenesis in p53-inactivated liver cancer. Mechanistically, we discovered that Il34 is a gene transcriptionally repressed by p53, and p53 loss resulted in IL-34 secretion by CSCs. IL-34 induced CD36-mediated elevations in fatty acid oxidative metabolism to drive M2-like polarization of foam-like tumor-associated macrophages (TAMs). These IL-34-orchestrated TAMs suppressed CD8+ T cell-mediated antitumor immunity to promote immune escape. Blockade of the IL-34-CD36 axis elicited antitumor immunity and synergized with anti-PD-1 immunotherapy, leading to a complete response. Our findings reveal the underlying mechanism of p53 modulation of the tumor immune microenvironment and provide a potential target for immunotherapy of cancer with p53 inactivation.


Asunto(s)
Interleucinas , Escape del Tumor , Microambiente Tumoral , Proteína p53 Supresora de Tumor , Macrófagos Asociados a Tumores , Animales , Humanos , Ratones , Antígenos CD36/metabolismo , Antígenos CD36/genética , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Reprogramación Celular/inmunología , Reprogramación Celular/genética , Inmunoterapia/métodos , Interleucinas/metabolismo , Interleucinas/inmunología , Neoplasias Hepáticas/inmunología , Ratones Endogámicos C57BL , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/metabolismo , Escape del Tumor/inmunología , Microambiente Tumoral/inmunología , Proteína p53 Supresora de Tumor/metabolismo , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo
3.
Front Immunol ; 15: 1424237, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39192979

RESUMEN

Triple-negative breast cancer (TNBC) has become a thorny problem in the treatment of breast cancer because of its high invasiveness, metastasis and recurrence. Although immunotherapy has made important progress in TNBC, immune escape caused by many factors, especially metabolic reprogramming, is still the bottleneck of TNBC immunotherapy. Regrettably, the mechanisms responsible for immune escape remain poorly understood. Exploring the mechanism of TNBC immune escape at the metabolic level provides a target and direction for follow-up targeting or immunotherapy. In this review, we focus on the mechanism that TNBC affects immune cells and interstitial cells through hypoxia, glucose metabolism, lipid metabolism and amino acid metabolism, and changes tumor metabolism and tumor microenvironment. This will help to find new targets and strategies for TNBC immunotherapy.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Escape del Tumor , Microambiente Tumoral , Humanos , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/metabolismo , Microambiente Tumoral/inmunología , Femenino , Animales , Inmunoterapia/métodos , Metabolismo de los Lípidos/inmunología , Reprogramación Celular/inmunología , Reprogramación Metabólica
4.
J Clin Invest ; 134(18)2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39052353

RESUMEN

Epithelial barriers are programmed for defense and repair but are also the site of long-term structural remodeling and disease. In general, this paradigm features epithelial stem cells (ESCs) that are called on to regenerate damaged tissues but can also be reprogrammed for detrimental remodeling. Here we identified a Wfdc21-dependent monocyte-derived dendritic cell (moDC) population that functioned as an early sentinel niche for basal ESC reprogramming in mouse models of epithelial injury after respiratory viral infection. Niche function depended on moDC delivery of ligand GPNMB to the basal ESC receptor CD44 so that properly timed antibody blockade of ligand or receptor provided long-lasting correction of reprogramming and broad disease phenotypes. These same control points worked directly in mouse and human basal ESC organoids. Together, the findings identify a mechanism to explain and modify what is otherwise a stereotyped but sometimes detrimental response to epithelial injury.


Asunto(s)
Reprogramación Celular , Animales , Ratones , Humanos , Reprogramación Celular/inmunología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Dendríticas/inmunología , Receptores de Hialuranos/metabolismo , Receptores de Hialuranos/inmunología , Receptores de Hialuranos/genética , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Nicho de Células Madre/inmunología , Células Madre/inmunología , Células Madre/metabolismo , Enfermedades Pulmonares/inmunología , Enfermedades Pulmonares/patología , Enfermedades Pulmonares/metabolismo
5.
Cancer Lett ; 597: 217076, 2024 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-38906524

RESUMEN

Understanding of the metabolic reprogramming has revolutionized our insights into tumor progression and potential treatment. This review concentrates on the aberrant metabolic pathways in cancer cells within the tumor microenvironment (TME). Cancer cells differ from normal cells in their metabolic processing of glucose, amino acids, and lipids in order to adapt to heightened biosynthetic and energy needs. These metabolic shifts, which crucially alter lactic acid, amino acid and lipid metabolism, affect not only tumor cell proliferation but also TME dynamics. This review also explores the reprogramming of various immune cells in the TME. From a therapeutic standpoint, targeting these metabolic alterations represents a novel cancer treatment strategy. This review also discusses approaches targeting the regulation of metabolism of different nutrients in tumor cells and influencing the tumor microenvironment to enhance the immune response. In summary, this review summarizes metabolic reprogramming in cancer and its potential as a target for innovative therapeutic strategies, offering fresh perspectives on cancer treatment.


Asunto(s)
Neoplasias , Microambiente Tumoral , Humanos , Microambiente Tumoral/inmunología , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/tratamiento farmacológico , Reprogramación Celular/inmunología , Animales , Metabolismo Energético/efectos de los fármacos , Metabolismo de los Lípidos , Reprogramación Metabólica
6.
Clin Immunol ; 265: 110287, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38909973

RESUMEN

LL37 alone and in complex with self-DNA triggers inflammatory responses in myeloid cells and plays a crucial role in the development of systemic autoimmune diseases, like psoriasis and systemic lupus erythematosus. We demonstrated that LL37/self-DNA complexes induce long-term metabolic and epigenetic changes in monocytes, enhancing their responsiveness to subsequent stimuli. Monocytes trained with LL37/self-DNA complexes and those derived from psoriatic patients exhibited heightened glycolytic and oxidative phosphorylation rates, elevated release of proinflammatory cytokines, and affected naïve CD4+ T cells. Additionally, KDM6A/B, a demethylase of lysine 27 on histone 3, was upregulated in psoriatic monocytes and monocytes treated with LL37/self-DNA complexes. Inhibition of KDM6A/B reversed the trained immune phenotype by reducing proinflammatory cytokine production, metabolic activity, and the induction of IL-17-producing T cells by LL37/self-DNA-treated monocytes. Our findings highlight the role of LL37/self-DNA-induced innate immune memory in psoriasis pathogenesis, uncovering its impact on monocyte and T cell dynamics.


Asunto(s)
Péptidos Catiónicos Antimicrobianos , Catelicidinas , ADN , Monocitos , Psoriasis , Humanos , Monocitos/inmunología , Monocitos/metabolismo , Psoriasis/inmunología , ADN/inmunología , ADN/metabolismo , Péptidos Catiónicos Antimicrobianos/metabolismo , Histona Demetilasas/metabolismo , Histona Demetilasas/genética , Linfocitos T CD4-Positivos/inmunología , Reprogramación Celular/inmunología , Citocinas/metabolismo , Citocinas/inmunología , Inmunidad Innata , Masculino , Epigénesis Genética , Femenino , Memoria Inmunológica , Histona Demetilasas con Dominio de Jumonji/metabolismo , Histona Demetilasas con Dominio de Jumonji/genética , Interleucina-17/metabolismo , Interleucina-17/inmunología , Células Cultivadas
7.
Gene ; 925: 148601, 2024 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-38788817

RESUMEN

Tumor-derived exosomes (TDEs), as topologies of tumor cells, not only carry biological information from the mother, but also act as messengers for cellular communication. It has been demonstrated that TDEs play a key role in inducing an immunosuppressive tumor microenvironment (TME). They can reprogram immune cells indirectly or directly by delivering inhibitory proteins, cytokines, RNA and other substances. They not only inhibit the maturation and function of dendritic cells (DCs) and natural killer (NK) cells, but also remodel M2 macrophages and inhibit T cell infiltration to promote immunosuppression and create a favorable ecological niche for tumor growth, invasion and metastasis. Based on the specificity of TDEs, targeting TDEs has become a new strategy to monitor tumor progression and enhance treatment efficacy. This paper reviews the intricate molecular mechanisms underlying the immunosuppressive effects induced by TDEs to establish a theoretical foundation for cancer therapy. Additionally, the challenges of TDEs as a novel approach to tumor treatment are discussed.


Asunto(s)
Exosomas , Neoplasias , Microambiente Tumoral , Exosomas/inmunología , Exosomas/metabolismo , Humanos , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Microambiente Tumoral/inmunología , Animales , Células Dendríticas/inmunología , Reprogramación Celular/inmunología , Células Asesinas Naturales/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Comunicación Celular/inmunología
8.
Immunol Lett ; 267: 106864, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38705481

RESUMEN

Tumor-associated myeloid cells, including macrophages and myeloid-derived suppressor cells, can be highly prevalent in solid tumors and play a significant role in the development of the tumor. Therefore, myeloid cells are being considered potential targets for cancer immunotherapies. In this review, we focused on strategies aimed at targeting tumor-associated macrophages (TAMs). Most strategies were studied preclinically but we also included a limited number of clinical studies based on these strategies. We describe possible underlying mechanisms and discuss future challenges and prospects.


Asunto(s)
Inmunoterapia , Neoplasias , Microambiente Tumoral , Macrófagos Asociados a Tumores , Humanos , Neoplasias/terapia , Neoplasias/inmunología , Inmunoterapia/métodos , Animales , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo , Microambiente Tumoral/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Reprogramación Celular/inmunología , Activación de Macrófagos/inmunología
10.
Immunopharmacol Immunotoxicol ; 46(3): 330-340, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38478467

RESUMEN

Rheumatoid arthritis (RA) is a persistent autoimmune condition characterized by ongoing inflammation primarily affecting the synovial joint. This inflammation typically arises from an increase in immune cells such as neutrophils, macrophages, and T cells (TC). TC is recognized as a major player in RA pathogenesis. The involvement of HLA-DRB1 and PTPN-2 among RA patients confirms the TC involvement in RA. Metabolism of TC is maintained by various other factors like cytokines, mitochondrial proteins & other metabolites. Different TC subtypes utilize different metabolic pathways like glycolysis, oxidative phosphorylation and fatty acid oxidation for their activation from naive TC (T0). Although all subsets of TC are not deleterious for synovium, some subsets of TC are involved in joint repair using their anti-inflammatory properties. Hence artificially reprogramming of TC subset by interfering with their metabolic status poised a hope in future to design new molecules against RA.


Asunto(s)
Artritis Reumatoide , Humanos , Artritis Reumatoide/inmunología , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo , Animales , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/efectos de los fármacos , Reprogramación Celular/efectos de los fármacos , Reprogramación Celular/inmunología
11.
Int Immunol ; 36(6): 261-278, 2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38364321

RESUMEN

Adoptive cell therapy (ACT) is an immunotherapeutic approach that involves isolating T cells from a patient, culturing them ex vivo, then reinfusing the cells back into the patient. Although this strategy has shown remarkable efficacy in hematological malignancies, the solid-tumour microenvironment (TME) has presented serious challenges for therapy efficacy. Particularly, the TME has immunosuppressive signalling and presents a metabolically challenging environment that leads to T-cell suppression. T-cell metabolism is an expanding field of research with a focus on understanding its inherent link to T-cell function. Here, we review the current model of T-cell metabolism from naïve cells through effector and memory life stages, as well as updates to the model from recent literature. These models of metabolism have provided us with the tools and understanding to explore T-cell metabolic and mitochondrial insufficiency in the TME. We discuss manipulations that can be made to these mitochondrial and metabolic pathways to enhance the persistence of infused T cells, overcome the metabolically challenging TME and improve the efficacy of therapy in ACT models. Further understanding and investigation of the impact of metabolic pathways on T-cell performance could contribute to improving therapy efficacy for patients.


Asunto(s)
Inmunoterapia Adoptiva , Linfocitos T , Humanos , Inmunoterapia Adoptiva/métodos , Animales , Linfocitos T/inmunología , Microambiente Tumoral/inmunología , Reprogramación Celular/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Mitocondrias/metabolismo , Mitocondrias/inmunología
12.
Mol Ther ; 32(9): 2835-2855, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-38273655

RESUMEN

The last decade has seen rapid development in the field of cellular immunotherapy, particularly in regard to chimeric antigen receptor (CAR)-modified T cells. However, challenges, such as severe treatment-related toxicities and inconsistent quality of autologous products, have hindered the broader use of CAR-T cell therapy, highlighting the need to explore alternative immune cells for cancer targeting. In this regard, natural killer (NK) cells have been extensively studied in cellular immunotherapy and were found to exert cytotoxic effects without being restricted by human leukocyte antigen and have a lower risk of causing graft-versus-host disease; making them favorable for the development of readily available "off-the-shelf" products. Clinical trials utilizing unedited NK cells or reprogrammed NK cells have shown early signs of their effectiveness against tumors. However, limitations, including limited in vivo persistence and expansion potential, remained. To enhance the antitumor function of NK cells, advanced gene-editing technologies and combination approaches have been explored. In this review, we summarize current clinical trials of antitumor NK cell therapy, provide an overview of innovative strategies for reprogramming NK cells, which include improvements in persistence, cytotoxicity, trafficking and the ability to counteract the immunosuppressive tumor microenvironment, and also discuss some potential combination therapies.


Asunto(s)
Inmunoterapia Adoptiva , Células Asesinas Naturales , Neoplasias , Microambiente Tumoral , Humanos , Células Asesinas Naturales/inmunología , Neoplasias/terapia , Neoplasias/inmunología , Inmunoterapia Adoptiva/métodos , Animales , Microambiente Tumoral/inmunología , Edición Génica , Reprogramación Celular/inmunología , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/genética , Ensayos Clínicos como Asunto
13.
Science ; 383(6679): eadf6493, 2024 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-38207030

RESUMEN

Neutrophils are increasingly recognized as key players in the tumor immune response and are associated with poor clinical outcomes. Despite recent advances characterizing the diversity of neutrophil states in cancer, common trajectories and mechanisms governing the ontogeny and relationship between these neutrophil states remain undefined. Here, we demonstrate that immature and mature neutrophils that enter tumors undergo irreversible epigenetic, transcriptional, and proteomic modifications to converge into a distinct, terminally differentiated dcTRAIL-R1+ state. Reprogrammed dcTRAIL-R1+ neutrophils predominantly localize to a glycolytic and hypoxic niche at the tumor core and exert pro-angiogenic function that favors tumor growth. We found similar trajectories in neutrophils across multiple tumor types and in humans, suggesting that targeting this program may provide a means of enhancing certain cancer immunotherapies.


Asunto(s)
Reprogramación Celular , Neoplasias , Neovascularización Patológica , Neutrófilos , Humanos , Neoplasias/irrigación sanguínea , Neoplasias/inmunología , Neutrófilos/inmunología , Proteómica , Reprogramación Celular/genética , Reprogramación Celular/inmunología , Neovascularización Patológica/genética , Neovascularización Patológica/inmunología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Epigénesis Genética , Hipoxia , Transcripción Genética
14.
Clin Transl Med ; 11(12): e634, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34965016

RESUMEN

BACKGROUND: Although microbioa-based therapies have shown putative effects on the treatment of non-alcoholic fatty liver disease (NAFLD), it is not clear how microbiota-derived metabolites contribute to the prevention of NAFLD. We explored the metabolomic signature of Lactobacillus lactis and Pediococcus pentosaceus in NAFLD mice and its association in NAFLD patients. METHODS: We used Western diet-induced NAFLD mice, and L. lactis and P. pentosaceus were administered to animals in the drinking water at a concentration of 109 CFU/g for 8 weeks. NAFLD severity was determined based on liver/body weight, pathology and biochemistry markers. Caecal samples were collected for the metagenomics by 16S rRNA sequencing. Metabolite profiles were obtained from caecum, liver and serum. Human stool samples (healthy control [n = 22] and NAFLD patients [n = 23]) were collected to investigate clinical reproducibility for microbiota-derived metabolites signature and metabolomics biomarker. RESULTS: L. lactis and P. pentosaceus supplementation effectively normalized weight ratio, NAFLD activity score, biochemical markers, cytokines and gut-tight junction. While faecal microbiota varied according to the different treatments, key metabolic features including short chain fatty acids (SCFAs), bile acids (BAs) and tryptophan metabolites were analogously restored by both probiotic supplementations. The protective effects of indole compounds were validated with in vitro and in vivo models, including anti-inflammatory effects. The metabolomic signatures were replicated in NAFLD patients, accompanied by the comparable levels of Firmicutes/Bacteroidetes ratio, which was significantly higher (4.3) compared with control (0.6). Besides, the consequent biomarker panel with six stool metabolites (indole, BAs, and SCFAs) showed 0.922 (area under the curve) in the diagnosis of NAFLD. CONCLUSIONS: NAFLD progression was robustly associated with metabolic dys-regulations in the SCFAs, bile acid and indole compounds, and NAFLD can be accurately diagnosed using the metabolites. L. lactis and P. pentosaceus ameliorate NAFLD progression by modulating gut metagenomic and metabolic environment, particularly tryptophan pathway, of the gut-liver axis.


Asunto(s)
Reprogramación Celular/inmunología , Microbioma Gastrointestinal/inmunología , Lactobacillus/metabolismo , Metaboloma/inmunología , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Pediococcus pentosaceus/metabolismo , Animales , Benzofuranos/metabolismo , Reprogramación Celular/fisiología , Dieta Occidental/efectos adversos , Modelos Animales de Enfermedad , Heces/microbiología , Microbioma Gastrointestinal/fisiología , Lactobacillus/patogenicidad , Metaboloma/fisiología , Ratones , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Pediococcus pentosaceus/patogenicidad , Quinolinas/metabolismo
15.
Int J Mol Sci ; 22(19)2021 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-34638609

RESUMEN

Immune escape is one of the hallmarks of cancer. While metabolic reprogramming provides survival advantage to tumor cancer cells, accumulating data also suggest such metabolic rewiring directly affects the activation, differentiation and function of immune cells, particularly in the tumor microenvironment. Understanding how metabolic reprogramming affects both tumor and immune cells, as well as their interplay, is therefore critical to better modulate tumor immune microenvironment in the era of cancer immunotherapy. In this review, we discuss alterations in several essential metabolic pathways in both tumor and key immune cells, provide evidence on their dynamic interaction, and propose innovative strategies to improve cancer immunotherapy via the modulation of metabolic pathways.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/terapia , Animales , Reprogramación Celular/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Progresión de la Enfermedad , Humanos , Inmunoterapia/tendencias , Macrófagos/inmunología , Macrófagos/metabolismo , Redes y Vías Metabólicas , Neoplasias/inmunología , Neoplasias/metabolismo , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Microambiente Tumoral/inmunología
16.
Immunity ; 54(9): 2024-2041.e8, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34473957

RESUMEN

Sepsis results in elevated adenosine in circulation. Extracellular adenosine triggers immunosuppressive signaling via the A2a receptor (A2aR). Sepsis survivors develop persistent immunosuppression with increased risk of recurrent infections. We utilized the cecal ligation and puncture (CLP) model of sepsis and subsequent infection to assess the role of adenosine in post-sepsis immune suppression. A2aR-deficient mice showed improved resistance to post-sepsis infections. Sepsis expanded a subset of CD39hi B cells and elevated extracellular adenosine, which was absent in mice lacking CD39-expressing B cells. Sepsis-surviving B cell-deficient mice were more resistant to secondary infections. Mechanistically, metabolic reprogramming of septic B cells increased production of ATP, which was converted into adenosine by CD39 on plasmablasts. Adenosine signaling via A2aR impaired macrophage bactericidal activity and enhanced interleukin-10 production. Septic individuals exhibited expanded CD39hi plasmablasts and adenosine accumulation. Our study reveals CD39hi plasmablasts and adenosine as important drivers of sepsis-induced immunosuppression with relevance in human disease.


Asunto(s)
Adenosina/inmunología , Antígenos CD/inmunología , Apirasa/inmunología , Tolerancia Inmunológica/inmunología , Macrófagos/inmunología , Células Plasmáticas/inmunología , Sepsis/inmunología , Adenosina/metabolismo , Animales , Antígenos CD/metabolismo , Apirasa/metabolismo , Reprogramación Celular/inmunología , Macrófagos/metabolismo , Ratones , Células Plasmáticas/metabolismo , Receptor de Adenosina A2A/inmunología , Receptor de Adenosina A2A/metabolismo , Sepsis/metabolismo
17.
Front Immunol ; 12: 706583, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34489958

RESUMEN

The burgeoning field of innate immune training, also called trained immunity, has given immunologists new insights into the role of innate responses in protection against infection and in modulating inflammation. Moreover, it has led to a paradigm shift in the way we think about immune memory and the interplay between innate and adaptive immune systems in conferring immunity against pathogens. Trained immunity is the term used to describe the medium-term epigenetic and metabolic reprogramming of innate immune cells in peripheral tissues or in the bone marrow stem cell niche. It is elicited by an initial challenge, followed by a significant period of rest that results in an altered response to a subsequent, unrelated challenge. Trained immunity can be associated with increased production of proinflammatory mediators, such as IL-1ß, TNF and IL-6, and increased expression of markers on innate immune cells associated with antigen presentation to T cells. The microenvironment created by trained innate immune cells during the secondary challenge may have profound effects on T cell responses, such as altering the differentiation, polarisation and function of T cell subtypes, including Th17 cells. In addition, the Th1 cytokine IFN-γ plays a critical role in establishing trained immunity. In this review, we discuss the evidence that trained immunity impacts on or can be impacted by T cells. Understanding the interplay between innate immune training and how it effects adaptive immunity will give insights into how this phenomenon may affect the development or progression of disease and how it could be exploited for therapeutic interventions or to enhance vaccine efficacy.


Asunto(s)
Inmunidad Adaptativa/inmunología , Inmunidad Innata/inmunología , Memoria Inmunológica/inmunología , Linfocitos T/inmunología , Animales , Reprogramación Celular/inmunología , Epigénesis Genética/inmunología , Humanos
18.
Mol Ther ; 29(11): 3192-3204, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34563675

RESUMEN

Cell therapies based on reprogrammed adaptive immune cells have great potential as "living drugs." As first demonstrated clinically for engineered chimeric antigen receptor (CAR) T cells, the ability of such cells to undergo clonal expansion in response to an antigen promotes both self-renewal and self-regulation in vivo. B cells also have the potential to be developed as immune cell therapies, but engineering their specificity and functionality is more challenging than for T cells. In part, this is due to the complexity of the immunoglobulin (Ig) locus, as well as the requirement for regulated expression of both cell surface B cell receptor and secreted antibody isoforms, in order to fully recapitulate the features of natural antibody production. Recent advances in genome editing are now allowing reprogramming of B cells by site-specific engineering of the Ig locus with preformed antibodies. In this review, we discuss the potential of engineered B cells as a cell therapy, the challenges involved in editing the Ig locus and the advances that are making this possible, and envision future directions for this emerging field of immune cell engineering.


Asunto(s)
Linfocitos B/metabolismo , Sistemas CRISPR-Cas , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Edición Génica , Terapia Genética/métodos , Inmunoterapia/métodos , Animales , Anticuerpos/genética , Anticuerpos/inmunología , Linfocitos B/inmunología , Ingeniería Celular , Reprogramación Celular/genética , Reprogramación Celular/inmunología , Regulación de la Expresión Génica , Ingeniería Genética , Humanos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
19.
Front Immunol ; 12: 714822, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34367185

RESUMEN

Advances in understanding how cancer cells interact with the immune system allowed the development of immunotherapeutic strategies, harnessing patients' immune system to fight cancer. Dendritic cell-based vaccines are being explored to reactivate anti-tumor adaptive immunity. Immune checkpoint inhibitors and chimeric antigen receptor T-cells (CAR T) were however the main approaches that catapulted the therapeutic success of immunotherapy. Despite their success across a broad range of human cancers, many challenges remain for basic understanding and clinical progress as only a minority of patients benefit from immunotherapy. In addition, cellular immunotherapies face important limitations imposed by the availability and quality of immune cells isolated from donors. Cell fate reprogramming is offering interesting alternatives to meet these challenges. Induced pluripotent stem cell (iPSC) technology not only enables studying immune cell specification but also serves as a platform for the differentiation of a myriad of clinically useful immune cells including T-cells, NK cells, or monocytes at scale. Moreover, the utilization of iPSCs allows introduction of genetic modifications and generation of T/NK cells with enhanced anti-tumor properties. Immune cells, such as macrophages and dendritic cells, can also be generated by direct cellular reprogramming employing lineage-specific master regulators bypassing the pluripotent stage. Thus, the cellular reprogramming toolbox is now providing the means to address the potential of patient-tailored immune cell types for cancer immunotherapy. In parallel, development of viral vectors for gene delivery has opened the door for in vivo reprogramming in regenerative medicine, an elegant strategy circumventing the current limitations of in vitro cell manipulation. An analogous paradigm has been recently developed in cancer immunotherapy by the generation of CAR T-cells in vivo. These new ideas on endogenous reprogramming, cross-fertilized from the fields of regenerative medicine and gene therapy, are opening exciting avenues for direct modulation of immune or tumor cells in situ, widening our strategies to remove cancer immunotherapy roadblocks. Here, we review current strategies for cancer immunotherapy, summarize technologies for generation of immune cells by cell fate reprogramming as well as highlight the future potential of inducing these unique cell identities in vivo, providing new and exciting tools for the fast-paced field of cancer immunotherapy.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Reprogramación Celular/genética , Reprogramación Celular/inmunología , Inmunoterapia , Terapia Molecular Dirigida , Neoplasias/etiología , Neoplasias/terapia , Animales , Antineoplásicos Inmunológicos/farmacología , Vacunas contra el Cáncer/uso terapéutico , Técnicas de Reprogramación Celular , Terapia Combinada , Manejo de la Enfermedad , Ingeniería Genética , Terapia Genética , Humanos , Inmunoterapia/efectos adversos , Inmunoterapia/métodos , Inmunoterapia Adoptiva , Terapia Molecular Dirigida/métodos
20.
Front Immunol ; 12: 717421, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34394124

RESUMEN

Regulatory T-cells (Tregs) are important for maintaining self-tolerance and tissue homeostasis. The functional plasticity of Tregs is a key feature of this lineage, as it allows them to adapt to different microenvironments, adopt transcriptional programs reflective of their environments and tailor their suppressive capacity in a context-dependent fashion. Tregs, particularly effector Tregs (eTregs), are abundant in many types of tumors. However, the functional and transcriptional plasticity of eTregs in tumors remain largely to be explored. Although depletion or inhibition of systemic Tregs can enhance anti-tumor responses, autoimmune sequelae have diminished the enthusiasm for such approaches. A more effective approach should specifically target intratumoral Tregs or subvert local Treg-mediated suppression. This mini-review will discuss the reported mechanisms by which the stability and suppressive function of tumoral Tregs are modulated, with the focus on eTregs and a subset of eTregs, follicular regulatory T (TFR) cells, and how to harness this knowledge for the future development of new effective cancer immunotherapies that selectively target the tumor local response while sparing the systemic side effects.


Asunto(s)
Reprogramación Celular , Susceptibilidad a Enfermedades , Neoplasias/etiología , Neoplasias/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Biomarcadores , Linaje de la Célula , Reprogramación Celular/genética , Reprogramación Celular/inmunología , Humanos , Neoplasias/patología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA