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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 22(6): 746-756, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34031618

RESUMEN

T cell exhaustion presents one of the major hurdles to cancer immunotherapy. Among exhausted CD8+ tumor-infiltrating lymphocytes, the terminally exhausted subset contributes directly to tumor cell killing owing to its cytotoxic effector function. However, this subset does not respond to immune checkpoint blockades and is difficult to be reinvigorated with restored proliferative capacity. Here, we show that a half-life-extended interleukin-10-Fc fusion protein directly and potently enhanced expansion and effector function of terminally exhausted CD8+ tumor-infiltrating lymphocytes by promoting oxidative phosphorylation, a process that was independent of the progenitor exhausted T cells. Interleukin-10-Fc was a safe and highly efficient metabolic intervention that synergized with adoptive T cell transfer immunotherapy, leading to eradication of established solid tumors and durable cures in the majority of treated mice. These findings show that metabolic reprogramming by upregulating mitochondrial pyruvate carrier-dependent oxidative phosphorylation can revitalize terminally exhausted T cells and enhance the response to cancer immunotherapy.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Interleucina-10/farmacología , Neoplasias/terapia , Fosforilación Oxidativa/efectos de los fármacos , Linfocitos T Citotóxicos/efectos de los fármacos , Animales , Proteínas de Transporte de Anión/genética , Proteínas de Transporte de Anión/metabolismo , Línea Celular Tumoral , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Células HEK293 , Semivida , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Fragmentos Fc de Inmunoglobulinas/farmacología , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Interleucina-10/uso terapéutico , Ratones , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Neoplasias/inmunología , Neoplasias/patología , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Receptores de Interleucina-10/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/uso terapéutico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo
2.
Nature ; 621(7980): 849-856, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37730993

RESUMEN

Protective immunity against pathogens or cancer is mediated by the activation and clonal expansion of antigen-specific naive T cells into effector T cells. To sustain their rapid proliferation and effector functions, naive T cells switch their quiescent metabolism to an anabolic metabolism through increased levels of aerobic glycolysis, but also through mitochondrial metabolism and oxidative phosphorylation, generating energy and signalling molecules1-3. However, how that metabolic rewiring drives and defines the differentiation of T cells remains unclear. Here we show that proliferating effector CD8+ T cells reductively carboxylate glutamine through the mitochondrial enzyme isocitrate dehydrogenase 2 (IDH2). Notably, deletion of the gene encoding IDH2 does not impair the proliferation of T cells nor their effector function, but promotes the differentiation of memory CD8+ T cells. Accordingly, inhibiting IDH2 during ex vivo manufacturing of chimeric antigen receptor (CAR) T cells induces features of memory T cells and enhances antitumour activity in melanoma, leukaemia and multiple myeloma. Mechanistically, inhibition of IDH2 activates compensating metabolic pathways that cause a disequilibrium in metabolites regulating histone-modifying enzymes, and this maintains chromatin accessibility at genes that are required for the differentiation of memory T cells. These findings show that reductive carboxylation in CD8+ T cells is dispensable for their effector response and proliferation, but that it mainly produces a pattern of metabolites that epigenetically locks CD8+ T cells into a terminal effector differentiation program. Blocking this metabolic route allows the increased formation of memory T cells, which could be exploited to optimize the therapeutic efficacy of CAR T cells.


Asunto(s)
Linfocitos T CD8-positivos , Activación de Linfocitos , Diferenciación Celular/genética , Ciclo del Ácido Cítrico , Fosforilación Oxidativa , Memoria Inmunológica/genética
3.
Immunity ; 49(6): 987-989, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30566885

RESUMEN

How macrophages convey extracellular signals by bridging metabolism and functions remains unclear. In this issue of Immunity, Sanin et al. (2018) report that prostaglandin E2 (PGE2) treatment in interleukin-4-activated macrophages suppresses mitochondrial membrane potential to control voltage-regulated genes involved in proliferation and immune responses.


Asunto(s)
Dinoprostona , Macrófagos , Expresión Génica , Potencial de la Membrana Mitocondrial
4.
Nature ; 479(7371): 122-6, 2011 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-21983962

RESUMEN

PHD2 serves as an oxygen sensor that rescues blood supply by regulating vessel formation and shape in case of oxygen shortage. However, it is unknown whether PHD2 can influence arteriogenesis. Here we studied the role of PHD2 in collateral artery growth by using hindlimb ischaemia as a model, a process that compensates for the lack of blood flow in case of major arterial occlusion. We show that Phd2 (also known as Egln1) haplodeficient (Phd2(+/-)) mice displayed preformed collateral arteries that preserved limb perfusion and prevented tissue necrosis in ischaemia. Improved arteriogenesis in Phd2(+/-) mice was due to an expansion of tissue-resident, M2-like macrophages and their increased release of arteriogenic factors, leading to enhanced smooth muscle cell (SMC) recruitment and growth. Both chronic and acute deletion of one Phd2 allele in macrophages was sufficient to skew their polarization towards a pro-arteriogenic phenotype. Mechanistically, collateral vessel preconditioning relied on the activation of canonical NF-κB pathway in Phd2(+/-) macrophages. These results unravel how PHD2 regulates arteriogenesis and artery homeostasis by controlling a specific differentiation state in macrophages and suggest new treatment options for ischaemic disorders.


Asunto(s)
Arterias/crecimiento & desarrollo , Extremidades/irrigación sanguínea , Isquemia/prevención & control , Macrófagos/metabolismo , Procolágeno-Prolina Dioxigenasa/deficiencia , Procolágeno-Prolina Dioxigenasa/metabolismo , Alelos , Animales , Modelos Animales de Enfermedad , Extremidades/patología , Femenino , Heterocigoto , Homeostasis , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Isquemia/patología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Miocitos del Músculo Liso/citología , FN-kappa B/metabolismo , Necrosis , Fenotipo , Procolágeno-Prolina Dioxigenasa/genética
5.
Gut ; 65(6): 990-1000, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-25814648

RESUMEN

OBJECTIVE: Cancer immunology is a growing field of research whose aim is to develop innovative therapies and diagnostic tests. Starting from the hypothesis that immune cells promptly respond to harmful stimuli, we used peripheral blood monocytes in order to characterise a distinct gene expression profile and to evaluate its potential as a candidate diagnostic biomarker in patients with colorectal cancer (CRC), a still unmet clinical need. DESIGN: We performed a case-control study including 360 peripheral blood monocyte samples from four European oncological centres and defined a gene expression profile specific to CRC. The robustness of the genetic profile and disease specificity were assessed in an independent setting. RESULTS: This screen returned 43 putative diagnostic markers, which we refined and validated in the confirmative multicentric analysis to 23 genes with outstanding diagnostic accuracy (area under the curve (AUC)=0.99 (0.99 to 1.00), Se=100.0% (100.0% to 100.0%), Sp=92.9% (78.6% to 100.0%) in multiple-gene receiver operating characteristic analysis). The diagnostic accuracy was robustly maintained in prospectively collected independent samples (AUC=0.95 (0.85 to 1.00), Se=92.6% (81.5% to 100.0%), Sp=92.3% (76.9% to 100.0%). This monocyte signature was expressed at early disease onset, remained robust over the course of disease progression, and was specific for the monocytic fraction of mononuclear cells. The gene modulation was induced specifically by soluble factors derived from transformed colon epithelium in comparison to normal colon or other cancer histotypes. Moreover, expression changes were plastic and reversible, as they were abrogated upon withdrawal of these tumour-released factors. Consistently, the modified set of genes reverted to normal expression upon curative treatment and was specific for CRC. CONCLUSIONS: Our study is the first to demonstrate monocyte plasticity in response to tumour-released soluble factors. The identified distinct signature in tumour-educated monocytes might be used as a candidate biomarker in CRC diagnosis and harbours the potential for disease follow-up and therapeutic monitoring.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Colorrectales/diagnóstico , Perfilación de la Expresión Génica , Monocitos , Anciano , Bélgica , Estudios de Casos y Controles , Neoplasias Colorrectales/sangre , Detección Precoz del Cáncer , Unión Europea , Femenino , Estudios de Seguimiento , Humanos , Técnicas In Vitro , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Valor Predictivo de las Pruebas , Sensibilidad y Especificidad
6.
Nat Biotechnol ; 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38168996

RESUMEN

The success of chimeric antigen receptor (CAR) T cell therapy in treating several hematopoietic malignancies has been difficult to replicate in solid tumors, in part because of T cell exhaustion and eventually dysfunction. To counter T cell dysfunction in the tumor microenvironment, we metabolically armored CAR T cells by engineering them to secrete interleukin-10 (IL-10). We show that IL-10 CAR T cells preserve intact mitochondrial structure and function in the tumor microenvironment and increase oxidative phosphorylation in a mitochondrial pyruvate carrier-dependent manner. IL-10 secretion promoted proliferation and effector function of CAR T cells, leading to complete regression of established solid tumors and metastatic cancers across several cancer types in syngeneic and xenograft mouse models, including colon cancer, breast cancer, melanoma and pancreatic cancer. IL-10 CAR T cells also induced stem cell-like memory responses in lymphoid organs that imparted durable protection against tumor rechallenge. Our results establish a generalizable approach to counter CAR T cell dysfunction through metabolic armoring, leading to solid tumor eradication and long-lasting immune protection.

7.
Nat Cancer ; 5(2): 240-261, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37996514

RESUMEN

Dendritic cells (DCs) are antigen-presenting myeloid cells that regulate T cell activation, trafficking and function. Monocyte-derived DCs pulsed with tumor antigens have been tested extensively for therapeutic vaccination in cancer, with mixed clinical results. Here, we present a cell-therapy platform based on mouse or human DC progenitors (DCPs) engineered to produce two immunostimulatory cytokines, IL-12 and FLT3L. Cytokine-armed DCPs differentiated into conventional type-I DCs (cDC1) and suppressed tumor growth, including melanoma and autochthonous liver models, without the need for antigen loading or myeloablative host conditioning. Tumor response involved synergy between IL-12 and FLT3L and was associated with natural killer and T cell infiltration and activation, M1-like macrophage programming and ischemic tumor necrosis. Antitumor immunity was dependent on endogenous cDC1 expansion and interferon-γ signaling but did not require CD8+ T cell cytotoxicity. Cytokine-armed DCPs synergized effectively with anti-GD2 chimeric-antigen receptor (CAR) T cells in eradicating intracranial gliomas in mice, illustrating their potential in combination therapies.


Asunto(s)
Citocinas , Neoplasias , Humanos , Ratones , Animales , Inmunoterapia , Células Dendríticas , Neoplasias/terapia , Interleucina-12
8.
Cancer Res Commun ; 3(4): 576-591, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37066023

RESUMEN

PI3K delta (PI3Kδ) inhibitors are used to treat lymphomas but safety concerns and limited target selectivity curbed their clinical usefulness. PI3Kδ inhibition in solid tumors has recently emerged as a potential novel anticancer therapy through the modulation of T-cell responses and direct antitumor activity. Here we report the exploration of IOA-244/MSC2360844, a first-in-class non-ATP-competitive PI3Kδ inhibitor, for the treatment of solid tumors. We confirm IOA-244's selectivity as tested against a large set of kinases, enzymes, and receptors. IOA-244 inhibits the in vitro growth of lymphoma cells and its activity correlates with the expression levels of PIK3CD, suggesting cancer cell-intrinsic effects of IOA-244. Importantly, IOA-244 inhibits regulatory T cell proliferation while having limited antiproliferative effects on conventional CD4+ T cells and no effect on CD8+ T cells. Instead, treatment of CD8 T cells with IOA-244 during activation, favors the differentiation of memory-like, long-lived CD8, known to have increased antitumor capacity. These data highlight immune-modulatory properties that can be exploited in solid tumors. In CT26 colorectal and Lewis lung carcinoma lung cancer models, IOA-244 sensitized the tumors to anti-PD-1 (programmed cell death protein 1) treatment, with similar activity in the Pan-02 pancreatic and A20 lymphoma syngeneic mouse models. IOA-244 reshaped the balance of tumor-infiltrating cells, favoring infiltration of CD8 and natural killer cells, while decreasing suppressive immune cells. IOA-244 presented no detectable safety concerns in animal studies and is currently in clinical phase Ib/II investigation in solid and hematologic tumors. Significance: IOA-244 is a first-in-class non-ATP-competitive, PI3Kδ inhibitor with direct antitumor in vitro activity correlated with PI3Kδ expression. The ability to modulate T cells, in vivo antitumor activity in various models with limited toxicity in animal studies provides the rationale for the ongoing trials in patients with solid tumors and hematologic cancers.


Asunto(s)
Linfoma , Neoplasias , Ratones , Animales , Linfocitos T CD8-positivos , Fosfatidilinositol 3-Quinasas , Neoplasias/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Linfoma/tratamiento farmacológico , Tolerancia Inmunológica
9.
Cell Metab ; 34(5): 731-746.e9, 2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35452600

RESUMEN

Glycolysis, including both lactate fermentation and pyruvate oxidation, orchestrates CD8+ T cell differentiation. However, how mitochondrial pyruvate metabolism and uptake controlled by the mitochondrial pyruvate carrier (MPC) impact T cell function and fate remains elusive. We found that genetic deletion of MPC drives CD8+ T cell differentiation toward a memory phenotype. Metabolic flexibility induced by MPC inhibition facilitated acetyl-coenzyme-A production by glutamine and fatty acid oxidation that results in enhanced histone acetylation and chromatin accessibility on pro-memory genes. However, in the tumor microenvironment, MPC is essential for sustaining lactate oxidation to support CD8+ T cell antitumor function. We further revealed that chimeric antigen receptor (CAR) T cell manufacturing with an MPC inhibitor imprinted a memory phenotype and demonstrated that infusing MPC inhibitor-conditioned CAR T cells resulted in superior and long-lasting antitumor activity. Altogether, we uncover that mitochondrial pyruvate uptake instructs metabolic flexibility for guiding T cell differentiation and antitumor responses.


Asunto(s)
Células T de Memoria , Transportadores de Ácidos Monocarboxílicos , Lactatos , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Ácido Pirúvico/metabolismo
10.
Front Immunol ; 13: 1011858, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36275777

RESUMEN

Autologous T cells expressing the Chimeric Antigen Receptor (CAR) have been approved as advanced therapy medicinal products (ATMPs) against several hematological malignancies. However, the generation of patient-specific CAR-T products delays treatment and precludes standardization. Allogeneic off-the-shelf CAR-T cells are an alternative to simplify this complex and time-consuming process. Here we investigated safety and efficacy of knocking out the TCR molecule in ARI-0001 CAR-T cells, a second generation αCD19 CAR approved by the Spanish Agency of Medicines and Medical Devices (AEMPS) under the Hospital Exemption for treatment of patients older than 25 years with Relapsed/Refractory acute B cell lymphoblastic leukemia (B-ALL). We first analyzed the efficacy and safety issues that arise during disruption of the TCR gene using CRISPR/Cas9. We have shown that edition of TRAC locus in T cells using CRISPR as ribonuleorproteins allows a highly efficient TCR disruption (over 80%) without significant alterations on T cells phenotype and with an increased percentage of energetic mitochondria. However, we also found that efficient TCRKO can lead to on-target large and medium size deletions, indicating a potential safety risk of this procedure that needs monitoring. Importantly, TCR edition of ARI-0001 efficiently prevented allogeneic responses and did not detectably alter their phenotype, while maintaining a similar anti-tumor activity ex vivo and in vivo compared to unedited ARI-0001 CAR-T cells. In summary, we showed here that, although there are still some risks of genotoxicity due to genome editing, disruption of the TCR is a feasible strategy for the generation of functional allogeneic ARI-0001 CAR-T cells. We propose to further validate this protocol for the treatment of patients that do not fit the requirements for standard autologous CAR-T cells administration.


Asunto(s)
Linfoma de Células B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Receptores Quiméricos de Antígenos , Humanos , Receptores Quiméricos de Antígenos/genética , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Linfocitos T , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Linfoma de Células B/etiología
11.
Cell Mol Immunol ; 18(7): 1761-1771, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-32055005

RESUMEN

Memory CD8 T cells can provide long-term protection against tumors, which depends on their enhanced proliferative capacity, self-renewal and unique metabolic rewiring to sustain cellular fitness. Specifically, memory CD8 T cells engage oxidative phosphorylation and fatty acid oxidation to fulfill their metabolic demands. In contrast, tumor-infiltrating lymphocytes (TILs) display severe metabolic defects, which may underlie their functional decline. Here, we show that overexpression of proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), the master regulator of mitochondrial biogenesis (MB), favors CD8 T cell central memory formation rather than resident memory generation. PGC-1α-overexpressing CD8 T cells persist and mediate more robust recall responses to bacterial infection or peptide vaccination. Importantly, CD8 T cells with enhanced PGC-1α expression provide stronger antitumor immunity in a mouse melanoma model. Moreover, TILs overexpressing PGC-1α maintain higher mitochondrial activity and improved expansion when rechallenged in a tumor-free host. Altogether, our findings indicate that enforcing mitochondrial biogenesis promotes CD8 T cell memory formation, metabolic fitness, and antitumor immunity in vivo.


Asunto(s)
Vacunas contra el Cáncer , Animales , Linfocitos T CD8-positivos/metabolismo , Ratones , Mitocondrias/metabolismo , Biogénesis de Organelos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Vacunas de Subunidad
12.
Methods Enzymol ; 631: 77-90, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31948568

RESUMEN

The formation of memory CD8+ T cells is crucial for host protection upon reencounter of the same pathogen. Moreover, long-lasting anti-tumoral effects of immunomodulatory drugs largely depend on the induction of immunological memory. While the transcriptional processes behind memory T cell differentiation have been described in detail, it is only recently becoming clear that memory T cell formation and maintenance are tightly controlled by specific metabolic pathways. Therefore, metabolic engineering of CD8+ T cells in order to promote their memory formation represents a valuable strategy to improve vaccination efficacy and cancer immunotherapy. Here, we describe several mouse in vitro and in vivo assays that can be used to evaluate whether a specific metabolic pathway is involved in memory CD8+ T cell differentiation. To this end, a metabolic process can be tested by either genetic deletion or pharmaceutical inhibition/activation of the key enzyme involved. The in vitro assays might allow for rapid screening of multiple candidate pathways, while the in vivo assays are required to reliably determine the quality and functionality of the generated memory CD8+ T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citometría de Flujo/métodos , Memoria Inmunológica , Técnicas Inmunológicas/métodos , Activación de Linfocitos , Animales , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/fisiología , Diferenciación Celular , Línea Celular Tumoral , Citocinas/análisis , Citocinas/metabolismo , Listeriosis , Ratones , Neoplasias/inmunología , Neoplasias/terapia
13.
Nat Rev Clin Oncol ; 16(7): 425-441, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30914826

RESUMEN

The development of immunotherapies over the past decade has resulted in a paradigm shift in the treatment of cancer. However, the majority of patients do not benefit from immunotherapy, presumably owing to insufficient reprogramming of the immunosuppressive tumour microenvironment (TME) and thus limited reinvigoration of antitumour immunity. Various metabolic machineries and nutrient-sensing mechanisms orchestrate the behaviour of immune cells in response to nutrient availability in the TME. Notably, tumour-infiltrating immune cells typically experience metabolic stress as a result of the dysregulated metabolic activity of tumour cells, leading to impaired antitumour immune responses. Moreover, the immune checkpoints that are often exploited by tumour cells to evade immunosurveillance have emerging roles in modulating the metabolic and functional activity of T cells. Thus, repurposing of drugs targeting cancer metabolism might synergistically enhance immunotherapy via metabolic reprogramming of the TME. In addition, interventions targeting the metabolic circuits that impede antitumour immunity have been developed, with several clinical trials underway. Herein, we discuss how these metabolic circuits regulate antitumour immunity and the possible approaches to targeting these pathways in the context of anticancer immunotherapy. We also describe hypothetical combination treatments that could be used to better unleash the potential of adoptive cell therapies by enhancing T cell metabolism.


Asunto(s)
Antineoplásicos/uso terapéutico , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Antineoplásicos/farmacología , Terapia Combinada , Reposicionamiento de Medicamentos , Sinergismo Farmacológico , Humanos , Redes y Vías Metabólicas/efectos de los fármacos , Neoplasias/inmunología , Microambiente Tumoral/efectos de los fármacos
14.
Cell Rep ; 21(10): 2842-2854, 2017 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-29212030

RESUMEN

Although it is well established that tumor-associated macrophages take part in each step of cancer progression, less is known about the distinct role of the so-called metastasis-associated macrophages (MAMs) at the metastatic site. Previous studies reported that Caveolin-1 (Cav1) has both tumor-promoting and tumor-suppressive functions. However, the role of Cav1 in bone-marrow-derived cells is unknown. Here, we describe Cav1 as an anti-metastatic regulator in mouse models of lung and breast cancer pulmonary metastasis. Among all the recruited inflammatory cell populations, we show that MAMs uniquely express abundant levels of Cav1. Using clodronate depletion of macrophages, we demonstrate that macrophage Cav1 signaling is critical for metastasis and not for primary tumor growth. In particular, Cav1 inhibition does not affect MAM recruitment to the metastatic site but, in turn, favors angiogenesis. We describe a mechanism by which Cav1 in MAMs specifically restrains vascular endothelial growth factor A/vascular endothelial growth factor receptor 1 (VEGF-A/VEGFR1) signaling and its downstream effectors, matrix metallopeptidase 9 (MMP9) and colony-stimulating factor 1 (CSF1).


Asunto(s)
Caveolina 1/metabolismo , Macrófagos/metabolismo , Animales , Caveolina 1/genética , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Noqueados , Metástasis de la Neoplasia , Neovascularización Patológica/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
15.
Cell Metab ; 24(5): 701-715, 2016 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-27773694

RESUMEN

Hypoxic tumor-associated macrophages (TAMs) acquire angiogenic and immunosuppressive properties. Yet it remains unknown if metabolic changes influence these functions. Here, we argue that hypoxic TAMs strongly upregulate the expression of REDD1, a negative regulator of mTOR. REDD1-mediated mTOR inhibition hinders glycolysis in TAMs and curtails their excessive angiogenic response, with consequent formation of abnormal blood vessels. Accordingly, REDD1 deficiency in TAMs leads to the formation of smoothly aligned, pericyte-covered, functional vessels, which prevents vessel leakiness, hypoxia, and metastases. Mechanistically, highly glycolytic REDD1-deficient TAMs outcompete endothelial cells for glucose usage that thwarts vascular hyperactivation and promotes the formation of quiescent vascular junctions. Tuning down glycolysis in REDD1 knockout TAMs re-establishes abnormal angiogenesis and metastases. On this basis, we prove that the anti-tumor effect of mTOR inhibitors is partly countered by the deleterious outcome of these drugs on TAMs. Our data provide a functional link between TAM metabolism and tumor angiogenesis.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Macrófagos/metabolismo , Morfogénesis , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Animales , Vasos Sanguíneos/metabolismo , Hipoxia de la Célula , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Eliminación de Gen , Glucosa/metabolismo , Glucólisis , Ratones Endogámicos C57BL , Ratones Noqueados , Metástasis de la Neoplasia , Neoplasias/patología , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/metabolismo , Regulación hacia Arriba
16.
Sci Rep ; 6: 33453, 2016 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-27634375

RESUMEN

Hepatic inflammation drives hepatic stellate cells (HSC), resulting in liver fibrosis. The Farnesoid-X receptor (FXR) antagonizes inflammation through NF-κB inhibition. We investigated preventive and therapeutic effects of FXR agonist obeticholic acid (OCA) on hepatic inflammation and fibrosis in toxic cirrhotic rats. Cirrhosis was induced by thioacetamide (TAA) intoxication. OCA was given during or after intoxication with vehicle-treated rats as controls. At sacrifice, fibrosis, hemodynamic and biochemical parameters were assessed. HSC activation, cell turn-over, hepatic NF-κB activation, pro-inflammatory and pro-fibrotic cytokines were determined. The effect of OCA was further evaluated in isolated HSC, Kupffer cells, hepatocytes and liver sinusoidal endothelial cells (LSEC). OCA decreased hepatic inflammation and fibrogenesis during TAA-administration and reversed fibrosis in established cirrhosis. Portal pressure decreased through reduced intrahepatic vascular resistance. This was paralleled by decreased expression of pro-fibrotic cytokines (transforming growth-factor ß, connective tissue growth factor, platelet-derived growth factor ß-receptor) as well as markers of hepatic cell turn-over, by blunting effects of pro-inflammatory cytokines (e.g. monocyte chemo-attractant protein-1). In vitro, OCA inhibited both LSEC and Kupffer cell activation; while HSC remained unaffected. This related to NF-κB inhibition via up-regulated IκBα. In conclusion, OCA inhibits hepatic inflammation in toxic cirrhotic rats resulting in decreased HSC activation and fibrosis.


Asunto(s)
Ácido Quenodesoxicólico/análogos & derivados , Inflamación/tratamiento farmacológico , Cirrosis Hepática/tratamiento farmacológico , Hígado/patología , Receptores Citoplasmáticos y Nucleares/agonistas , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Ácido Quenodesoxicólico/farmacología , Ácido Quenodesoxicólico/uso terapéutico , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Hemodinámica/efectos de los fármacos , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Inflamación/complicaciones , Inflamación/patología , Inflamación/fisiopatología , Mediadores de Inflamación/metabolismo , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/patología , Lipopolisacáridos/farmacología , Cirrosis Hepática/complicaciones , Cirrosis Hepática/patología , Cirrosis Hepática/fisiopatología , Masculino , Ratones , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B/metabolismo , Presión Portal/efectos de los fármacos , Ratas Wistar , Receptores Citoplasmáticos y Nucleares/metabolismo , Tioacetamida , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/efectos de los fármacos , Resistencia Vascular/efectos de los fármacos
17.
Cancer Cell ; 24(6): 695-709, 2013 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-24332039

RESUMEN

Recruitment of tumor-associated macrophages (TAMs) into avascular areas sustains tumor progression; however, the underlying guidance mechanisms are unknown. Here, we report that hypoxia-induced Semaphorin 3A (Sema3A) acts as an attractant for TAMs by triggering vascular endothelial growth factor receptor 1 phosphorylation through the associated holoreceptor, composed of Neuropilin-1 (Nrp1) and PlexinA1/PlexinA4. Importantly, whereas Nrp1 levels are downregulated in the hypoxic environment, Sema3A continues to regulate TAMs in an Nrp1-independent manner by eliciting PlexinA1/PlexinA4-mediated stop signals, which retain them inside the hypoxic niche. Consistently, gene deletion of Nrp1 in macrophages favors TAMs' entrapment in normoxic tumor regions, which abates their pro-angiogenic and immunosuppressive functions, hence inhibiting tumor growth and metastasis. This study shows that TAMs' heterogeneity depends on their localization, which is tightly controlled by Sema3A/Nrp1 signaling.


Asunto(s)
Macrófagos/fisiología , Neoplasias Experimentales/irrigación sanguínea , Neovascularización Patológica/prevención & control , Neuropilina-1/fisiología , Semaforina-3A/fisiología , Transducción de Señal/fisiología , Animales , Hipoxia de la Célula , Macrófagos/efectos de los fármacos , Ratones , Ratones Noqueados , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Neuropilina-1/antagonistas & inhibidores , Neuropilina-1/genética , Semaforina-3A/antagonistas & inhibidores
18.
EMBO Mol Med ; 5(6): 843-57, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23616286

RESUMEN

Occlusion of the main arterial route redirects blood flow to the collateral circulation. We previously reported that macrophages genetically modified to express low levels of prolyl hydroxylase domain protein 2 (PHD2) display an arteriogenic phenotype, which promotes the formation of collateral vessels and protects the skeletal muscle from ischaemic necrosis. However, the molecular mechanisms underlying this process are unknown. Here, we demonstrate that femoral artery occlusion induces a switch in macrophage phenotype through angiopoietin-1 (ANG1)-mediated Phd2 repression. ANG blockade by a soluble trap prevented the downregulation of Phd2 expression in macrophages and their phenotypic switch, thus inhibiting collateral growth. ANG1-dependent Phd2 repression initiated a feed-forward loop mediated by the induction of the ANG receptor TIE2 in macrophages. Gene silencing and cell depletion strategies demonstrate that TIE2 induction in macrophages is required to promote their proarteriogenic functions, enabling collateral vessel formation following arterial obstruction. These results indicate an indispensable role for TIE2 in sustaining in situ programming of macrophages to a proarteriogenic, M2-like phenotype, suggesting possible new venues for the treatment of ischaemic disorders.


Asunto(s)
Procolágeno-Prolina Dioxigenasa/metabolismo , Receptor TIE-2/metabolismo , Angiopoyetina 1/genética , Angiopoyetina 1/metabolismo , Angiopoyetina 2/genética , Angiopoyetina 2/metabolismo , Animales , Regulación hacia Abajo , Silenciador del Gen , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Isquemia/metabolismo , Isquemia/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , MicroARNs/metabolismo , Fenotipo , Procolágeno-Prolina Dioxigenasa/genética , Transducción de Señal
19.
Immunobiology ; 217(12): 1292-300, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22944250

RESUMEN

The vitamin D receptor (VDR) is a hormone nuclear receptor regulating bone and calcium homeostasis. Studies revealing the expression of VDR on immune cells point toward a role for VDR-dependent signaling pathways in immunity. Here we verified the ability of the natural VDR ligand, 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)) to interfere in inflammatory and T cell stimulatory capacity of macrophages, in particular within a chronic inflammatory disease features of experimental type 1 diabetes (T1D). We demonstrated that VDR is constitutively expressed in macrophages and both the levels of VDR and its downstream targets, are clearly induced by 1,25(OH)(2)D(3). In control mice, macrophage programming with 1,25(OH)(2)D(3) partially abrogated the activation-provoked expression of IL-12p40, TNFα and iNOS as well as the effector T cell-recruiting chemokines, CXCL9, CXCL10 and CXCL11. Targeting VDR signaling in macrophages counteracted their T-cell stimulatory ability despite essentially unaltered expression of antigen-presenting and costimulatory molecules. Furthermore, even in non-obese diabetic (NOD) mice, where macrophages/monocytes featured a heightened responsiveness toward danger signals and a superior T cell stimulatory capacity, 1,25(OH)(2)D(3) successfully curtailed these basic macrophage-mediated functions. Interestingly, the inhibitory action of the active compound was associated with an IL-10-dependent mechanism since 1,25(OH)(2)D(3)-treatment of IL-10-deficient macrophages failed to reproduce the characteristic repression on inflammatory mediators or T cell proliferation. Combined, these results highlight the possible therapeutic applicability of this natural immunomodulator, due to its ability to counteract macrophage inflammatory and T cell-activating pathways.


Asunto(s)
Calcitriol/inmunología , Inflamación/inmunología , Interleucina-10/inmunología , Macrófagos/inmunología , Linfocitos T/inmunología , Animales , Calcitriol/metabolismo , Células Cultivadas , Quimiocinas CXC/inmunología , Quimiocinas CXC/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Modelos Animales de Enfermedad , Femenino , Inflamación/metabolismo , Interleucina-10/metabolismo , Subunidad p40 de la Interleucina-12/inmunología , Subunidad p40 de la Interleucina-12/metabolismo , Activación de Linfocitos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Óxido Nítrico Sintasa de Tipo II/inmunología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Receptores de Calcitriol/inmunología , Receptores de Calcitriol/metabolismo , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
20.
Cancer Res ; 72(8): 1953-63, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22374984

RESUMEN

Histidine-rich glycoprotein (HRG) is a 75-kDa heparin-binding plasma protein implicated in the regulation of tumor growth and vascularization. In this study, we show that hrg(-/-) mice challenged with fibrosarcoma or pancreatic carcinoma grow larger tumors with increased metastatic properties. Compared with wild-type mice, fibrosarcomas in hrg(-/-) mice were more hypoxic, necrotic, and less perfused, indicating enhanced vessel abnormalization. HRG deficiency was associated with a suppressed antitumor immune response, with both increased infiltration of M2 marker-expressing macrophages and decreased infiltration of dendritic cells and cytotoxic T cells. Analysis of transcript expression in tumor-associated as well as peritoneal macrophages from hrg(-/-) mice revealed an increased expression of genes associated with a proangiogenic and immunoinhibitory phenotype. In accordance, expression arrays conducted on HRG-treated peritoneal macrophages showed induction of genes involved in extracellular matrix biology and immune responsiveness. In conclusion, our findings show that macrophages are a direct target of HRG. HRG loss influences macrophage gene regulation, leading to excessive stimulation of tumor angiogenesis, suppression of tumor immune response, and increased tumor growth and metastatic spread.


Asunto(s)
Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Neovascularización Patológica/patología , Proteínas/metabolismo , Escape del Tumor/fisiología , Animales , Humanos , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Metástasis de la Neoplasia , Neoplasias Experimentales/genética , Neovascularización Patológica/inmunología , Neovascularización Patológica/metabolismo , Proteínas/genética , Proteínas/inmunología , Microambiente Tumoral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA