Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nat Immunol ; 23(10): 1470-1483, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36138183

RESUMEN

Traditionally viewed as poorly plastic, neutrophils are now recognized as functionally diverse; however, the extent and determinants of neutrophil heterogeneity in humans remain unclear. We performed a comprehensive immunophenotypic and transcriptome analysis, at a bulk and single-cell level, of neutrophils from healthy donors and patients undergoing stress myelopoiesis upon exposure to growth factors, transplantation of hematopoietic stem cells (HSC-T), development of pancreatic cancer and viral infection. We uncover an extreme diversity of human neutrophils in vivo, reflecting the rates of cell mobilization, differentiation and exposure to environmental signals. Integrated control of developmental and inducible transcriptional programs linked flexible granulopoietic outputs with elicitation of stimulus-specific functional responses. In this context, we detected an acute interferon (IFN) response in the blood of patients receiving HSC-T that was mirrored by marked upregulation of IFN-stimulated genes in neutrophils but not in monocytes. Systematic characterization of human neutrophil plasticity may uncover clinically relevant biomarkers and support the development of diagnostic and therapeutic tools.


Asunto(s)
Mielopoyesis , Neutrófilos , Biomarcadores/metabolismo , Humanos , Interferones/genética , Interferones/metabolismo , Neutrófilos/metabolismo , Plásticos/metabolismo
2.
Immunity ; 55(6): 973-975, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35704999

RESUMEN

In this issue of Immunity, Eisenstein, Hiliard, et al., uncover a novel mechanism of some widely used non-steroidal anti-inflammatory drugs (NSAIDs): activation of the antioxidant transcription factor NRF2 in myeloid immune cells.


Asunto(s)
Antiinflamatorios no Esteroideos , Inflamación , Antiinflamatorios no Esteroideos/uso terapéutico , Humanos
3.
Immunity ; 54(8): 1665-1682.e14, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34129840

RESUMEN

Tight control of inflammatory gene expression by antagonistic environmental cues is key to ensure immune protection while preventing tissue damage. Prostaglandin E2 (PGE2) modulates macrophage activation during homeostasis and disease, but the underlying mechanisms remain incompletely characterized. Here we dissected the genomic properties of lipopolysaccharide (LPS)-induced genes whose expression is antagonized by PGE2. The latter molecule targeted a set of inflammatory gene enhancers that, already in unstimulated macrophages, displayed poorly permissive chromatin organization and were marked by the transcription factor myocyte enhancer factor 2A (MEF2A). Deletion of MEF2A phenocopied PGE2 treatment and abolished type I interferon (IFN I) induction upon exposure to innate immune stimuli. Mechanistically, PGE2 interfered with LPS-mediated activation of ERK5, a known transcriptional partner of MEF2. This study highlights principles of plasticity and adaptation in cells exposed to a complex environment and uncovers a transcriptional circuit for IFN I induction with relevance for infectious diseases or cancer.


Asunto(s)
Dinoprostona/inmunología , Interferón Tipo I/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Animales , Línea Celular , Células Cultivadas , Regulación de la Expresión Génica/inmunología , Humanos , Inflamación/genética , Inflamación/inmunología , Interferón Tipo I/biosíntesis , Lipopolisacáridos , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 7 Activada por Mitógenos/metabolismo
4.
Nature ; 623(7986): 415-422, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37914939

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease with high resistance to therapies1. Inflammatory and immunomodulatory signals co-exist in the pancreatic tumour microenvironment, leading to dysregulated repair and cytotoxic responses. Tumour-associated macrophages (TAMs) have key roles in PDAC2, but their diversity has prevented therapeutic exploitation. Here we combined single-cell and spatial genomics with functional experiments to unravel macrophage functions in pancreatic cancer. We uncovered an inflammatory loop between tumour cells and interleukin-1ß (IL-1ß)-expressing TAMs, a subset of macrophages elicited by a local synergy between prostaglandin E2 (PGE2) and tumour necrosis factor (TNF). Physical proximity with IL-1ß+ TAMs was associated with inflammatory reprogramming and acquisition of pathogenic properties by a subset of PDAC cells. This occurrence was an early event in pancreatic tumorigenesis and led to persistent transcriptional changes associated with disease progression and poor outcomes for patients. Blocking PGE2 or IL-1ß activity elicited TAM reprogramming and antagonized tumour cell-intrinsic and -extrinsic inflammation, leading to PDAC control in vivo. Targeting the PGE2-IL-1ß axis may enable preventive or therapeutic strategies for reprogramming of immune dynamics in pancreatic cancer.


Asunto(s)
Inflamación , Interleucina-1beta , Neoplasias Pancreáticas , Macrófagos Asociados a Tumores , Humanos , Carcinogénesis , Carcinoma Ductal Pancreático/complicaciones , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Dinoprostona/metabolismo , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Inflamación/complicaciones , Inflamación/inmunología , Inflamación/patología , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Neoplasias Pancreáticas/complicaciones , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Microambiente Tumoral , Factores de Necrosis Tumoral/metabolismo , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo , Macrófagos Asociados a Tumores/patología
5.
Immunol Rev ; 300(1): 220-236, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33565148

RESUMEN

Most, if not all, aspects of carcinogenesis are influenced by the tumor microenvironment (TME), a complex architecture of cells, matrix components, soluble signals, and their dynamic interactions in the context of physical traits of the tissue. Expanding application of technologies for high-dimensional analyses with single-cell resolution has begun to decipher the contributions of the immune system to cancer progression and its implications for therapy. In this review, we will discuss the multifaceted roles of tumor-associated macrophages and neutrophils, focusing on factors that subvert tissue immune homeostasis and offer therapeutic opportunities for TME reprogramming. By performing a critical analysis of available datasets, we elaborate on diversification mechanisms and unifying principles of myeloid cell heterogeneity in human tumors.


Asunto(s)
Neoplasias , Microambiente Tumoral , Carcinogénesis , Humanos , Células Mieloides , Neoplasias/terapia , Neutrófilos
6.
J Allergy Clin Immunol ; 142(5): 1605-1617.e4, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29447842

RESUMEN

BACKGROUND: Wiskott-Aldrich syndrome (WAS) is a rare primary immunodeficiency caused by mutations in Wiskott-Aldrich syndrome protein (WASp), a key regulator of cytoskeletal dynamics in hematopoietic cells. A high proportion of patients experience autoimmunity caused by a breakdown in T- and B-cell tolerance. Moreover, excessive production of type I interferon (IFN-I) by plasmacytoid dendritic cells (pDCs) contributes to autoimmune signs; however, the factors that trigger excessive innate activation have not been defined. OBJECTIVE: Neutrophil extracellular traps (NETs) emerged as major initiating factors in patients with diseases such as systemic lupus erythematosus and rheumatoid arthritis. In this study we explored the possible involvement of aberrant neutrophil functions in patients with WAS. METHODS: We evaluated the expression of a set of granulocyte genes associated with NETs in a cohort of patients with WAS and the presence of NET inducers in sera. Using a mouse model of WAS, we analyzed NET release by WASp-null neutrophils and evaluated the composition and homeostasis of neutrophils in vivo. By using depletion experiments, we assessed the effect of neutrophils in promoting inflammation and reactivity against autoantigens. RESULTS: Transcripts of genes encoding neutrophil enzymes and antimicrobial peptides were increased in granulocytes of patients with WAS, and serum-soluble factors triggered NET release. WASp-null neutrophils showed increased spontaneous NETosis, induced IFN-I production by pDCs, and activated B cells through B-cell activating factor. Consistently, their depletion abolished constitutive pDC activation, normalized circulating IFN-I levels, and, importantly, abolished production of autoantibodies directed against double-stranded DNA, nucleosomes, and myeloperoxidase. CONCLUSIONS: These findings reveal that neutrophils are involved in the pathogenic loop that causes excessive activation of innate cells and autoreactive B cells, thus identifying novel mechanisms that contribute to the autoimmunity of WAS.


Asunto(s)
Interferón Tipo I/inmunología , Neutrófilos/inmunología , Síndrome de Wiskott-Aldrich/inmunología , Adolescente , Adulto , Animales , Autoanticuerpos/inmunología , Linfocitos B/inmunología , Preescolar , Células Dendríticas/inmunología , Trampas Extracelulares , Femenino , Expresión Génica , Humanos , Lactante , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Síndrome de Wiskott-Aldrich/genética , Adulto Joven
7.
Blood ; 119(22): 5250-60, 2012 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-22504926

RESUMEN

The atypical chemokine receptor D6 is a decoy and scavenger receptor for most inflammatory CC chemokines and prevents the development of exacerbated inflammatory reactions. Here we report that mice lacking D6 expression in the nonhematopoietic compartment have a selective increase in the number of Ly6C(high) monocytes in the circulation and in secondary lymphoid tissues. Under inflammatory conditions, Ly6C(high) monocytes accumulate in increased number in secondary lymphoid organs of D6(-/-) mice in a CCR2-dependent manner. Ly6C(high) monocytes derived from D6(-/-) mice have enhanced immunosuppressive activity, inhibit the development of adaptive immune responses, and partially protect mice from the development of GVHD. Thus, control of CCR2 ligands by D6 regulates the traffic of Ly6C(high) monocytes and controls their immunosuppressive potential.


Asunto(s)
Inmunidad Adaptativa/fisiología , Antígenos Ly , Regulación de la Expresión Génica/inmunología , Tolerancia Inmunológica/fisiología , Monocitos/inmunología , Receptores de Quimiocina/inmunología , Animales , Regulación de la Expresión Génica/genética , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Inflamación/genética , Inflamación/inmunología , Ratones , Ratones Noqueados , Receptores CCR2/genética , Receptores CCR2/inmunología , Receptores de Quimiocina/genética
8.
Nat Commun ; 15(1): 2280, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38480738

RESUMEN

Cross-presentation by type 1 DCs (cDC1) is critical to induce and sustain antitumoral CD8 T cell responses to model antigens, in various tumor settings. However, the impact of cross-presenting cDC1 and the potential of DC-based therapies in tumors carrying varied levels of bona-fide neoantigens (neoAgs) remain unclear. Here we develop a hypermutated model of non-small cell lung cancer in female mice, encoding genuine MHC-I neoepitopes to study neoAgs-specific CD8 T cell responses in spontaneous settings and upon Flt3L + αCD40 (DC-therapy). We find that cDC1 are required to generate broad CD8 responses against a range of diverse neoAgs. DC-therapy promotes immunogenicity of weaker neoAgs and strongly inhibits the growth of high tumor-mutational burden (TMB) tumors. In contrast, low TMB tumors respond poorly to DC-therapy, generating mild CD8 T cell responses that are not sufficient to block progression. scRNA transcriptional analysis, immune profiling and functional assays unveil the changes induced by DC-therapy in lung tissues, which comprise accumulation of cDC1 with increased immunostimulatory properties and less exhausted effector CD8 T cells. We conclude that boosting cDC1 activity is critical to broaden the diversity of anti-tumoral CD8 T cell responses and to leverage neoAgs content for therapeutic advantage.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Femenino , Ratones , Animales , Células Dendríticas , Carcinoma de Pulmón de Células no Pequeñas/terapia , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/terapia , Neoplasias Pulmonares/metabolismo , Linfocitos T CD8-positivos , Reactividad Cruzada
9.
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
10.
Oncoimmunology ; 11(1): 2059876, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35402081

RESUMEN

Lung tumor-infiltrating neutrophils are known to support growth and dissemination of cancer cells and to suppress T cell responses. However, the precise impact of tissue neutrophils on programming and differentiation of anticancer CD8 T cells in vivo remains poorly understood. Here, we identified cancer cell-autonomous secretion of CXCL5 as sufficient to drive infiltration of mature, protumorigenic neutrophils in a mouse model of non-small cell lung cancer (NSCLC). Consistently, CXCL5 transcripts correlate with neutrophil density and poor prognosis in a large human lung adenocarcinoma compendium. CXCL5 genetic deletion, unlike antibody-mediated depletion, completely and selectively prevented neutrophils accumulation in lung tissues. Depletion of tumor-infiltrating neutrophils promoted expansion of tumor-specific CD8 T cells, differentiation into effector cells and acquisition of cytolytic functions. Transfer of effector CD8 T cells into neutrophil-rich tumors, inhibited IFN-ϒ production, indicating active suppression of effector functions. Importantly, blocking neutrophils infiltration in the lung, overcame resistance to checkpoint blockade. Hence, this study demonstrates that neutrophils curb acquisition of cytolytic functions in lung tumor tissues and suggests targeting of CXCL5 as a strategy to restore anti-tumoral T cell functions.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Animales , Linfocitos T CD8-positivos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Pulmón/patología , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Neutrófilos
11.
Cancer Immunol Res ; 10(11): 1340-1353, 2022 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-36122412

RESUMEN

TIM4 has previously been associated with antitumor immunity, yet the pattern of expression and the function of this receptor across human cancer tissues remain poorly explored. Here we combined extensive immunolabeling of human tissues with in silico analysis of pan-cancer transcriptomic data sets to explore the clinical significance of TIM4 expression. Our results unveil that TIM4 is expressed on a fraction of cavity macrophages (CATIM4+MΦ) of carcinoma patients. Moreover, we uncover a high expression of TIM4 on macrophages of the T-cell zone of the carcinoma-associated tertiary lymphoid structures (TLSTIM4+MΦ). In silico analysis of a pan-cancer data set revealed a positive correlation between TIM4 expression and markers of B cells, effector CD8+ T cells, and a 12-chemokine signature defining tertiary lymphoid structure. In addition, TLSTIM4+MΦ were enriched in cancers displaying microsatellite instability and high CD8+ T-cell infiltration, confirming their association with immune-reactive tumors. Both CATIM4+MΦ and TLSTIM4+MΦ express FOLR2, a marker of tissue-resident MΦ. However, CATIM4+MΦ had a higher expression of the immunosuppressive molecules TREM2, IL10, and TGFß as compared with TLSTIM4+MΦ. By analyzing a scRNA sequence data set of tumor-associated myeloid cells, we identified two TIM4+FOLR2+ clusters coherent with CATIM4+MΦ and TLSTIM4+MΦ. We defined specific gene signatures for each subset and found that the CATIM4+ MΦ signature was associated with worse patient survival. In contrast, TLSTIM4+MΦ gene signature positively correlates with a better prognosis. Together, these data illustrate that TIM4 marks two distinct macrophage populations with distinct phenotypes and tissue localization and that may have opposing roles in tumor immunity.


Asunto(s)
Carcinoma , Receptor 2 de Folato , Estructuras Linfoides Terciarias , Humanos , Macrófagos , Linfocitos T CD8-positivos , Quimiocinas/metabolismo , Carcinoma/metabolismo , Receptor 2 de Folato/metabolismo
12.
Nat Commun ; 12(1): 2237, 2021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33854047

RESUMEN

Acquisition of cell-associated tumor antigens by type 1 dendritic cells (cDC1) is essential to induce and sustain tumor specific CD8+ T cells via cross-presentation. Here we show that capture and engulfment of cell associated antigens by tissue resident lung cDC1 is inhibited during progression of mouse lung tumors. Mechanistically, loss of phagocytosis is linked to tumor-mediated downregulation of the phosphatidylserine receptor TIM4, that is highly expressed in normal lung resident cDC1. TIM4 receptor blockade and conditional cDC1 deletion impair activation of tumor specific CD8+ T cells and promote tumor progression. In human lung adenocarcinomas, TIM4 transcripts increase the prognostic value of a cDC1 signature and predict responses to PD-1 treatment. Thus, TIM4 on lung resident cDC1 contributes to immune surveillance and its expression is suppressed in advanced tumors.


Asunto(s)
Antígenos de Neoplasias/inmunología , Células Dendríticas/inmunología , Neoplasias Pulmonares/inmunología , Proteínas de la Membrana/inmunología , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Animales , Antígenos de Neoplasias/genética , Linfocitos T CD8-positivos/inmunología , Reactividad Cruzada , Humanos , Vigilancia Inmunológica , Pulmón/inmunología , Neoplasias Pulmonares/genética , Proteínas de la Membrana/genética , Ratones
13.
JCI Insight ; 5(17)2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32721945

RESUMEN

Dysregulated sensing of self-nucleic acid is a leading cause of autoimmunity in multifactorial and monogenic diseases. Mutations in Wiskott-Aldrich syndrome protein (WASp), a key regulator of cytoskeletal dynamics in immune cells, cause autoimmune manifestations and increased production of type I IFNs by innate cells. Here we show that immune complexes of self-DNA and autoantibodies (DNA-ICs) contribute to elevated IFN levels via activation of the cGAS/STING pathway of cytosolic sensing. Mechanistically, lack of endosomal F-actin nucleation by WASp caused a delay in endolysosomal maturation and prolonged the transit time of ingested DNA-ICs. Stalling in maturation-defective organelles facilitated leakage of DNA-ICs into the cytosol, promoting activation of the TBK1/STING pathway. Genetic deletion of STING and STING and cGAS chemical inhibitors abolished IFN production and rescued systemic activation of IFN-stimulated genes in vivo. These data unveil the contribution of cytosolic self-nucleic acid sensing in WAS and underscore the importance of WASp-mediated endosomal actin remodeling in preventing innate activation.


Asunto(s)
ADN/inmunología , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/metabolismo , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Actinas/metabolismo , Animales , Autoanticuerpos/inmunología , Células Cultivadas , Células Dendríticas/inmunología , Endosomas/metabolismo , Inmunidad Innata , Interferones/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/metabolismo
14.
Cancer Res ; 78(7): 1685-1699, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29363545

RESUMEN

Restoring antigen presentation for efficient and durable activation of tumor-specific CD8+ T-cell responses is pivotal to immunotherapy, yet the mechanisms that cause subversion of dendritic cell (DC) functions are not entirely understood, limiting the development of targeted approaches. In this study, we show that bona fide DCs resident in lung tumor tissues or DCs exposed to factors derived from whole lung tumors become refractory to endosomal and cytosolic sensor stimulation and fail to secrete IL12 and IFNI. Tumor-conditioned DC exhibited downregulation of the SNARE VAMP3, a regulator of endosomes trafficking critical for cross-presentation of tumor antigens and DC-mediated tumor rejection. Dissection of cell-extrinsic suppressive pathways identified lactic acid in the tumor microenvironment as sufficient to inhibit type-I IFN downstream of TLR3 and STING. DC conditioning by lactate also impacted adaptive function, accelerating antigen degradation and impairing cross-presentation. Importantly, DCs conditioned by lactate failed to prime antitumor responses in vivo These findings provide a new mechanistic viewpoint to the concept of DC suppression and hold potential for future therapeutic approaches.Significance: These findings provide insight into the cell-intrinsic and cell-extrinsic mechanisms that cause loss of presentation of tumor-specific antigens in lung cancer tissues. Cancer Res; 78(7); 1685-99. ©2018 AACR.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Neoplasias Pulmonares/inmunología , Proteínas de Transporte de Membrana/biosíntesis , Animales , Línea Celular Tumoral , Medios de Cultivo Condicionados/metabolismo , Regulación hacia Abajo , Endosomas/metabolismo , Inmunoterapia , Interferón Tipo I/antagonistas & inhibidores , Ácido Láctico/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas SNARE/biosíntesis , Microambiente Tumoral/inmunología , Proteína 3 de Membrana Asociada a Vesículas/biosíntesis
15.
Nat Commun ; 9(1): 676, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29445158

RESUMEN

Atypical chemokine receptors (ACKRs) are regulators of leukocyte traffic, inflammation, and immunity. ACKR2 is a scavenger for most inflammatory CC chemokines and is a negative regulator of inflammation. Here we report that ACKR2 is expressed in hematopoietic precursors and downregulated during myeloid differentiation. Genetic inactivation of ACKR2 results in increased levels of inflammatory chemokine receptors and release from the bone marrow of neutrophils with increased anti-metastatic activity. In a model of NeuT-driven primary mammary carcinogenesis ACKR2 deficiency is associated with increased primary tumor growth and protection against metastasis. ACKR2 deficiency results in neutrophil-mediated protection against metastasis in mice orthotopically transplanted with 4T1 mammary carcinoma and intravenously injected with B16F10 melanoma cell lines. Thus, ACKR2 is a key regulator (checkpoint) of mouse myeloid differentiation and function and its targeting unleashes the anti-metastatic activity of neutrophils in mice.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Neoplasias Experimentales/metabolismo , Neutrófilos/metabolismo , Receptores de Quimiocina/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular Tumoral , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/metabolismo , Metástasis de la Neoplasia , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Receptores de Quimiocina/genética
16.
Methods Mol Biol ; 1393: 87-96, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27033218

RESUMEN

Chemokines are a large family of secreted cytokines whose main function is to mediate leukocyte directional migration. Most cancers contain chemokines and express chemokine receptors as a consequence of the activity of deregulated transcription factors or tumor-suppressor genes. Indeed chemokines expression at the tumor site dictates leukocyte infiltration and angiogenesis, while chemokine receptors expression by tumor cells promotes their growth and matastatization. Chemokines also have several indirect effects on tumor growth and are a relevant element in the cancer-related inflammation. In this chapter we will describe technical approaches available to study the role of chemokines in leukocyte infiltration and tumor metastatization in murine tumor models.


Asunto(s)
Adenocarcinoma/inmunología , Quimiocinas/fisiología , Neoplasias Pulmonares/inmunología , Adenocarcinoma/secundario , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Neoplasias Pulmonares/secundario , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias
17.
EBioMedicine ; 11: 165-172, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27475897

RESUMEN

Laser therapy, recently renamed as photobiomodulation, stands as a promising supportive treatment for oral mucositis induced by oncological therapies. However, its mechanisms of action and, more importantly, its safety in cancer patients, are still unclear. Here we explored the anti-cancer effect of 3 laser protocols, set at the most commonly used wavelengths, in B16F10 melanoma and oral carcinogenesis mouse models. While laser light increased cell metabolism in cultured cells, the in vivo outcome was reduced tumor progression. This striking, unexpected result, was paralleled by the recruitment of immune cells, in particular T lymphocytes and dendritic cells, which secreted type I interferons. Laser light also reduced the number of highly angiogenic macrophages within the tumor mass and promoted vessel normalization, an emerging strategy to control tumor progression. Collectively, these results set photobiomodulation as a safety procedure in oncological patients and open the way to its innovative use for cancer therapy.


Asunto(s)
Vigilancia Inmunológica/efectos de la radiación , Terapia por Láser , Neoplasias/inmunología , Neoplasias/patología , Neovascularización Patológica , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de la radiación , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/efectos de la radiación , Modelos Animales de Enfermedad , Femenino , Humanos , Interferones/biosíntesis , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Melanoma Experimental , Ratones , Ratones Noqueados , Invasividad Neoplásica , Neoplasias/genética , Neoplasias/terapia , Neovascularización Patológica/terapia , Carga Tumoral/efectos de la radiación
18.
Immunobiology ; 220(2): 249-53, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25454487

RESUMEN

Myeloid cells are key elements of the cancer-related inflammation with the potential to support not only tumor growth but also invasion and metastasis. Tumor-derived factors affect myeloid cell differentiation inducing a phenotype that supports tumor growth, inducing immunosuppression, angiogenesis and tissue remodeling. Soluble mediators, produced at primary tumor site, can also act in a remote mode inducing the release from bone marrow of myeloid cells that have immunosuppressive activities in tumor-draining lymphoid organs and can predispose to colonization when migrate to metastatic organs. We will here review current knowledge on the contribution of tumor-derived signals that affect polarized activation of myeloid cells, their bone marrow release and recruitment to metastatic sites with a particular focus on the role of chemokines.


Asunto(s)
Inflamación/inmunología , Células Mieloides/inmunología , Neoplasias/inmunología , Neoplasias/patología , Animales , Médula Ósea/inmunología , Médula Ósea/metabolismo , Movimiento Celular/inmunología , Humanos , Inflamación/metabolismo , Inflamación/patología , Células Mieloides/metabolismo , Metástasis de la Neoplasia , Neoplasias/metabolismo , Microambiente Tumoral
19.
Oncoimmunology ; 3(12): e955337, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25964861

RESUMEN

Inflammatory chemokines are instrumental players in cancer-related inflammation contributing to numerous steps during tumor progression. In Kaposi's sarcoma, we have found that downregulation of the atypical chemokine receptor 2 (ACKR2) by the KRAS/BRAF/ERK pathway profoundly affects the tumor microenvironment, unleashing accumulation of tumor-associated macrophages that sustains tumor growth. This discovery extends our understanding on the role of inflammatory chemokines in tumor biology and provides rationale for their therapeutic targeting.

20.
Cancer Immunol Res ; 2(7): 679-89, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24844911

RESUMEN

D6 is an atypical chemokine receptor acting as a decoy and scavenger for inflammatory CC chemokines expressed in lymphatic endothelial cells. Here, we report that D6 is expressed in Kaposi sarcoma (KS), a tumor ontogenetically related to the lymphatic endothelium. Both in human tumors and in an experimental model, D6 expression levels were inversely correlated with tumor aggressiveness and increased infiltration of proangiogenic macrophages. Inhibition of monocyte recruitment reduced the growth of tumors, while adoptive transfer of wild-type, but not CCR2(-/-) macrophages, increased the growth rate of D6-competent neoplasms. In the KS model with the B-Raf V600E-activating mutation, inhibition of B-Raf or the downstream ERK pathway induced D6 expression; in progressing human KS tumors, the activation of ERK correlates with reduced levels of D6 expression. These results indicate that activation of the K-Ras-B-Raf-ERK pathway during KS progression downregulates D6 expression, which unleashes chemokine-mediated macrophage recruitment and their acquisition of an M2-like phenotype supporting angiogenesis and tumor growth. Combined targeting of CCR2 and the ERK pathway should be considered as a therapeutic option for patients with KS.


Asunto(s)
Regulación hacia Abajo/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Receptores CCR10/biosíntesis , Sarcoma de Kaposi/inmunología , Animales , Citocinas/metabolismo , Progresión de la Enfermedad , Xenoinjertos , Humanos , Mediadores de Inflamación/metabolismo , Leucocitos/inmunología , Macrófagos/inmunología , Ratones Endogámicos C57BL , Ratones Desnudos , Trasplante de Neoplasias , Neovascularización Patológica/inmunología , Receptores CCR10/inmunología , Sarcoma de Kaposi/irrigación sanguínea , Sarcoma de Kaposi/patología , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Receptor de Quimiocina D6
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA