Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 158
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 49(3): 504-514.e4, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30231984

RESUMEN

The adaptor protein CARD9 links detection of fungi by surface receptors to the activation of the NF-κB pathway. Mice deficient in CARD9 exhibit dysbiosis and are more susceptible to colitis. Here we examined the impact of Card9 deficiency in the development of colitis-associated colon cancer (CAC). Treatment of Card9-/- mice with AOM-DSS resulted in increased tumor loads as compared to WT mice and in the accumulation of myeloid-derived suppressor cells (MDSCs) in tumor tissue. The impaired fungicidal functions of Card9-/- macrophages led to increased fungal loads and variation in the overall composition of the intestinal mycobiota, with a notable increase in C. tropicalis. Bone marrow cells incubated with C. tropicalis exhibited MDSC features and suppressive functions. Fluconazole treatment suppressed CAC in Card9-/- mice and was associated with decreased MDSC accumulation. The frequency of MDSCs in tumor tissues of colon cancer patients correlated positively with fungal burden, pointing to the relevance of this regulatory axis in human disease.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Colitis/inmunología , Neoplasias del Colon/inmunología , Disbiosis/inmunología , Microbioma Gastrointestinal/inmunología , Células Supresoras de Origen Mieloide/fisiología , Animales , Proteínas Adaptadoras de Señalización CARD/genética , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Colitis/inducido químicamente , Colitis/genética , Neoplasias del Colon/genética , Disbiosis/genética , Humanos , Interferón gamma/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Supresoras de Origen Mieloide/microbiología , Regiones Promotoras Genéticas/genética
2.
Nature ; 579(7798): 284-290, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32103175

RESUMEN

Cancer recurrence after surgery remains an unresolved clinical problem1-3. Myeloid cells derived from bone marrow contribute to the formation of the premetastatic microenvironment, which is required for disseminating tumour cells to engraft distant sites4-6. There are currently no effective interventions that prevent the formation of the premetastatic microenvironment6,7. Here we show that, after surgical removal of primary lung, breast and oesophageal cancers, low-dose adjuvant epigenetic therapy disrupts the premetastatic microenvironment and inhibits both the formation and growth of lung metastases through its selective effect on myeloid-derived suppressor cells (MDSCs). In mouse models of pulmonary metastases, MDSCs are key factors in the formation of the premetastatic microenvironment after resection of primary tumours. Adjuvant epigenetic therapy that uses low-dose DNA methyltransferase and histone deacetylase inhibitors, 5-azacytidine and entinostat, disrupts the premetastatic niche by inhibiting the trafficking of MDSCs through the downregulation of CCR2 and CXCR2, and by promoting MDSC differentiation into a more-interstitial macrophage-like phenotype. A decreased accumulation of MDSCs in the premetastatic lung produces longer periods of disease-free survival and increased overall survival, compared with chemotherapy. Our data demonstrate that, even after removal of the primary tumour, MDSCs contribute to the development of premetastatic niches and settlement of residual tumour cells. A combination of low-dose adjuvant epigenetic modifiers that disrupts this premetastatic microenvironment and inhibits metastases may permit an adjuvant approach to cancer therapy.


Asunto(s)
Epigénesis Genética , Terapia Genética , Células Supresoras de Origen Mieloide/fisiología , Neoplasias/terapia , Microambiente Tumoral , Animales , Azacitidina/farmacología , Benzamidas/farmacología , Diferenciación Celular , Movimiento Celular/efectos de los fármacos , Quimioterapia Adyuvante , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Ratones , Células Supresoras de Origen Mieloide/citología , Metástasis de la Neoplasia/terapia , Neoplasias/cirugía , Piridinas/farmacología , Receptores CCR2/genética , Receptores de Interleucina-8B/genética , Microambiente Tumoral/efectos de los fármacos
3.
EMBO J ; 38(11)2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31000523

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are immature myeloid cells with strong immunosuppressive activity that promote tumor growth. In this study, we describe a mechanism by which cancer cells control MDSCs in human cancers by upregulating TRF2, a protein required for telomere stability. Specifically, we showed that the TRF2 upregulation in cancer cells has extratelomeric roles in activating the expression of a network of genes involved in the biosynthesis of heparan sulfate proteoglycan, leading to profound changes in glycocalyx length and stiffness, as revealed by atomic force microscopy. This TRF2-dependent regulation facilitated the recruitment of MDSCs, their activation via the TLR2/MyD88/IL-6/STAT3 pathway leading to the inhibition of natural killer recruitment and cytotoxicity, and ultimately tumor progression and metastasis. The clinical relevance of these findings is supported by our analysis of cancer cohorts, which showed a correlation between high TRF2 expression and MDSC infiltration, which was inversely correlated with overall patient survival.


Asunto(s)
Glicocálix/metabolismo , Neoplasias/inmunología , Neoplasias/patología , Proteína 2 de Unión a Repeticiones Teloméricas/fisiología , Escape del Tumor/fisiología , Animales , Células Cultivadas , Femenino , Regulación Neoplásica de la Expresión Génica , Glicocálix/genética , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/fisiología , Células 3T3 NIH , Neoplasias/genética , Neoplasias/mortalidad , Telómero/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Escape del Tumor/genética
4.
Proc Natl Acad Sci U S A ; 117(33): 20159-20170, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32747553

RESUMEN

Although immune checkpoint blockade (ICB) therapy has revolutionized cancer treatment, many patients do not respond or develop resistance to ICB. N6 -methylation of adenosine (m6A) in RNA regulates many pathophysiological processes. Here, we show that deletion of the m6A demethylase Alkbh5 sensitized tumors to cancer immunotherapy. Alkbh5 has effects on m6A density and splicing events in tumors during ICB. Alkbh5 modulates Mct4/Slc16a3 expression and lactate content of the tumor microenvironment and the composition of tumor-infiltrating Treg and myeloid-derived suppressor cells. Importantly, a small-molecule Alkbh5 inhibitor enhanced the efficacy of cancer immunotherapy. Notably, the ALKBH5 gene mutation and expression status of melanoma patients correlate with their response to immunotherapy. Our results suggest that m6A demethylases in tumor cells contribute to the efficacy of immunotherapy and identify ALKBH5 as a potential therapeutic target to enhance immunotherapy outcome in melanoma, colorectal, and potentially other cancers.


Asunto(s)
Desmetilasa de ARN, Homólogo 5 de AlkB/metabolismo , Vacunas contra el Cáncer/inmunología , Lactatos/metabolismo , Melanoma/metabolismo , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T Reguladores/fisiología , Desmetilasa de ARN, Homólogo 5 de AlkB/genética , Anticuerpos , Citocinas/genética , Citocinas/metabolismo , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/terapia , Metiltransferasas/genética , Metiltransferasas/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Células Supresoras de Origen Mieloide/fisiología , Sitios de Empalme de ARN , Empalme del ARN , Simportadores/genética , Simportadores/metabolismo , Transcriptoma , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
Exp Cell Res ; 399(2): 112467, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33428904

RESUMEN

Tumor-associated macrophages (TAM), which are found in the tumor microenvironment of solid tumors, not only mediate cancer immune evasion but also promote tumor growth. The transcription factor NF-κB, which is a crucial link between inflammation and tumors, can accelerate tumor occurrence and development. NEMO, the regulatory subunit of the IKK complex, plays a pivotal role in activating the NF-κB signaling pathway. However, the function of myeloid NEMO in the tumor microenvironment remains unclear. Here, we found that conditional knockout of NEMO in myeloid cells promoted tumor growth in a transplanted cancer mouse model. In Nemofl/fl lyz-cre+/- mice, the deletion of Nemo in myeloid cells increased the recruitment of M2 macrophages and myeloid-derived suppressor cells (MDSCs) into the tumor, reduced the expression of apoptosis-related proteins, and upregulated the expression of the chemokine receptor CCR2, thereby promoting tumor growth in vivo. Then, we showed that blocking the MCP1-CCR2 pathway could inhibit tumor growth, especially in mice with myeloid NEMO deletion. In this study, we examined the mechanism of NEMO in myeloid cells and explored the role of NEMO in the prevention and treatment of cancer.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Células Mieloides/metabolismo , Escape del Tumor/genética , Animales , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Eliminación de Gen , Tolerancia Inmunológica/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/patología , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/fisiología , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Receptores CCR2/genética , Receptores CCR2/metabolismo , Transducción de Señal/genética , Células Tumorales Cultivadas , Escape del Tumor/inmunología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
6.
Immunol Rev ; 281(1): 57-61, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29247996

RESUMEN

Inflammation is an important component of the tumor microenvironment. IL-1 is an inflammatory cytokine which plays a key role in carcinogenesis and tumor progression. IL-1 is subject to regulation by components of the IL-1 and IL-1 receptor (ILR) families. Negative regulators include a decoy receptor (IL-1R2), receptor antagonists (IL-1Ra), IL-1R8, and anti-inflammatory IL-37. IL-1 acts at different levels in tumor initiation and progression, including driving chronic non-resolving inflammation, tumor angiogenesis, activation of the IL-17 pathway, induction of myeloid-derived suppressor cells (MDSC) and macrophage recruitment, invasion and metastasis. Based on initial clinical results, the translation potential of IL-1 targeting deserves extensive analysis.


Asunto(s)
Inmunoterapia/métodos , Interleucina-1/metabolismo , Células Supresoras de Origen Mieloide/fisiología , Neoplasias/inmunología , Animales , Carcinogénesis , Humanos , Inmunomodulación , Interleucina-17/metabolismo , Metástasis de la Neoplasia , Neovascularización Patológica , Receptores de Interleucina-1/metabolismo , Transducción de Señal , Microambiente Tumoral
7.
J Clin Immunol ; 41(3): 515-525, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33387156

RESUMEN

PURPOSE: The SARS-CoV-2 infection can lead to a severe acute respiratory distress syndrome (ARDS) with prolonged mechanical ventilation and high mortality rate. Interestingly, COVID-19-associated ARDS share biological and clinical features with sepsis-associated immunosuppression since lymphopenia and acquired infections associated with late mortality are frequently encountered. Mechanisms responsible for COVID-19-associated lymphopenia need to be explored since they could be responsible for delayed virus clearance and increased mortality rate among intensive care unit (ICU) patients. METHODS: A series of 26 clinically annotated COVID-19 patients were analyzed by thorough phenotypic and functional investigations at days 0, 4, and 7 after ICU admission. RESULTS: We revealed that, in the absence of any difference in demographic parameters nor medical history between the two groups, ARDS patients presented with an increased number of myeloid-derived suppressor cells (MDSC) and a decreased number of CD8pos effector memory cell compared to patients hospitalized for COVID-19 moderate pneumonia. Interestingly, COVID-19-related MDSC expansion was directly correlated to lymphopenia and enhanced arginase activity. Lastly, T cell proliferative capacity in vitro was significantly reduced among COVID-19 patients and could be restored through arginine supplementation. CONCLUSIONS: The present study reports a critical role for MDSC in COVID-19-associated ARDS. Our findings open the possibility of arginine supplementation as an adjuvant therapy for these ICU patients, aiming to reduce immunosuppression and help virus clearance, thereby decreasing the duration of mechanical ventilation, nosocomial infection acquisition, and mortality.


Asunto(s)
Arginina/metabolismo , COVID-19/complicaciones , Linfopenia/etiología , Células Supresoras de Origen Mieloide/fisiología , Síndrome de Dificultad Respiratoria/inmunología , SARS-CoV-2 , Anciano , Infección Hospitalaria/etiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/metabolismo , Índice de Severidad de la Enfermedad
8.
Cell Immunol ; 362: 104297, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33550187

RESUMEN

The challenge of distinguishing between changes attributable to ageing and those attributable to pathology is even greater for the immune system than for many other organs, and this is especially true for myeloid-derived suppressor cells (MDSCs). Hematopoiesis is different in older adults with a bias towards myelopoiesis, and older adults also manifest "inflammageing" exacerbated by disease and contributing to MDSC induction. Hence, at least in humans, one can only investigate MDSCs in the context of ageing and disease states, and not in the context of ageing processes per se. This contribution provides a brief overview of the literature on MDSCs and ageing in humans.


Asunto(s)
Envejecimiento/inmunología , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/fisiología , Envejecimiento/fisiología , Humanos
9.
Cell Immunol ; 362: 104301, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33588246

RESUMEN

Immuno checkpoint blockade (ICB) targeting the PD-1/PD-L1 axis is the main breakthrough for the treatment of several cancers. Nevertheless, not all patients benefit from this treatment and clinical response not always correlates with PD-L1 expression by tumor cells. The tumor microenvironment, including myeloid derived suppressor cells (MDSCs), can influence therapeutic resistance to ICB. MDSCs also express PD-L1, which contributes to their suppressive activity. Moreover, anticancer therapies including chemotherapy, radiotherapy, hormone- and targeted- therapies can modulate MDSCs recruitment, activity and PD-L1 expression. Such effects can be induced also by innovative anticancer treatments targeting metabolism and lifestyle. The outcome on cancer progression can be either positive or negative, depending on tumor type, treatment schedule and possible combination with ICB. Further studies are needed to better understand the effects of cancer therapies on the PD-1/PD-L1 axis, to identify patients that could benefit from combinatorial regimens including ICB or that rather should avoid it.


Asunto(s)
Antígeno B7-H1/metabolismo , Células Supresoras de Origen Mieloide/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Antígeno B7-H1/inmunología , Antígeno B7-H1/fisiología , Línea Celular Tumoral , Humanos , Inmunoterapia , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/fisiología , Neoplasias/inmunología , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/inmunología , Microambiente Tumoral
10.
Cell Immunol ; 361: 104280, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33445053

RESUMEN

Myeloid derived suppressor cells (MDSC) are a heterogenous population of immature myeloid cells that accumulate in tumor bearing host and migrate to lymphoid organs and tumor tissues. This process is controlled by a set of defined pro-inflammatory cytokines and chemokines, which are upregulated in malignancies. MDSC have strong immunosuppressive potential and constitute a major component of the tumor microenvironment (TME). Tumor cells take advantage of the suppressive mechanisms of MDSC to establish an immunosuppressive TME which inhibits antitumor immune responses thereby promoting cancer progression. An immunosuppressive TME acts as a significant barrier to immunotherapeutic interventions. Pre-clinical and clinical studies have demonstrated that enrichment and activation of MDSC is correlated with tumor progression, recurrence and metastasis. In this review we discuss the potential impact of MDSC on tumor progression and its role as a biomarker of prognostic significance in cancer with a special focus on hepatocellular cancer (HCC).


Asunto(s)
Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Neoplasias/inmunología , Animales , Biomarcadores de Tumor/inmunología , Carcinoma Hepatocelular/inmunología , Quimiocinas/inmunología , Citocinas/inmunología , Progresión de la Enfermedad , Humanos , Terapia de Inmunosupresión , Neoplasias Hepáticas/inmunología , Células Mieloides/inmunología , Células Supresoras de Origen Mieloide/fisiología , Recurrencia Local de Neoplasia/inmunología , Microambiente Tumoral/inmunología
11.
Cell Immunol ; 361: 104285, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33484943

RESUMEN

Myeloid derived suppressor cells (MDSCs) are a diverse collection of immune cells that suppress anti-tumor immune responses. Decreasing MDSCs accumulation in the tumor microenvironment could improve the anti-tumor immune response and improve immunotherapy. Here, we examine the impact of physiologically relevant thermal treatments on the accumulation of MDSCs in tumors in mice. We found that different temperature-based protocols, including 1) weekly whole-body hyperthermia, 2) housing mice at their thermoneutral temperature (TT, ~30 °C), and 3) housing mice at a subthermoneutral temperature (ST,~22 °C) while providing a localized heat source, each resulted in a reduction in MDSC accumulation and improved tumor growth control compared to control mice housed at ST, which is the standard, mandated housing temperature for laboratory mice. Additionally, we found that low dose ß-adrenergic receptor blocker (propranolol) therapy reduced MDSC accumulation and improved tumor growth control to a similar degree as the models that relieved cold stress. These results show that thermal treatments can decrease MDSC accumulation and tumor growth comparable to propranolol therapy.


Asunto(s)
Calor/uso terapéutico , Células Supresoras de Origen Mieloide/inmunología , Neoplasias/inmunología , Antagonistas Adrenérgicos beta/farmacología , Animales , Línea Celular Tumoral , Femenino , Respuesta al Choque Térmico/fisiología , Calefacción/métodos , Hipertermia Inducida/métodos , Inmunoterapia/métodos , Masculino , Ratones , Ratones Endogámicos BALB C , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/fisiología , Microambiente Tumoral/inmunología
12.
Cell Immunol ; 362: 104298, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33592541

RESUMEN

Myeloid derived suppressor cells (MDSCs) are a highly heterogeneous population of immature immune cells with immunosuppressive functions that are recruited to the tumor microenvironment (TME). MDSCs promote tumor growth and progression by inhibiting immune effector cell proliferation and function. MDSCs are affected by both novel anti-cancer therapies targeting the immune system to promote anti-tumor immunity, as well as by conventional treatments such as radiotherapy. Following radiotherapy, cytoplasmic double stranded DNA stimulates the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway, resulting in type I interferon production. Effectiveness of radiotherapy and cGAS/STING signaling are closely intertwined: activation of cGAS and STING is key to generate systemic anti-tumor immunity after irradiation. This review focuses on how radiotherapy and cGAS/STING signaling in MDSCs and/or tumor cells impact MDSC recruitment, expansion and function. The influence of conventional and ablative radiotherapy treatment schedules, inflammatory response following radiotherapy, and hypoxia are discussed as MDSC modulators.


Asunto(s)
Proteínas de la Membrana/metabolismo , Células Supresoras de Origen Mieloide/inmunología , Nucleotidiltransferasas/metabolismo , Humanos , Inmunidad Innata , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Proteínas de la Membrana/fisiología , Células Supresoras de Origen Mieloide/fisiología , Neoplasias/patología , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/fisiología , Radioterapia/métodos , Transducción de Señal/inmunología , Microambiente Tumoral/inmunología , Microambiente Tumoral/fisiología
13.
Cell Immunol ; 362: 104296, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33556903

RESUMEN

Efficient priming of anti-tumor T cells requires the uptake and presentation of tumor antigens by immunogenic dendritic cells (DCs) and occurs mainly in lymph nodes draining the tumor (tdLNs). However, tumors expand and activate myeloid-derived suppressor cells (MDSCs) that inhibit CTL functions by several mechanisms. While the immune-suppressive nature of the tumor microenvironment is largely documented, it is not known whether similar immune-suppressive mechanisms operate in the tdLNs. In this study, we analyzed MDSC characteristics within tdLNs. We show that, in a metastasis-free context, MO-MDSCs are the dominant MDSC population within tdLNs, that they are highly suppressive and that tumor proximity enhances their recruitment to tdLN via a CCR2/CCL2-dependent pathway. Altogether our results uncover a mechanism by which tumors evade the immune system that involves MDSC-mediated recruitment to the tdLN and the inhibition of T-cell activation even before reaching the highly immunosuppressive tumor microenvironment.


Asunto(s)
Células Supresoras de Origen Mieloide/metabolismo , Receptores CCR2/metabolismo , Microambiente Tumoral/inmunología , Animales , Línea Celular Tumoral , Femenino , Humanos , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/fisiología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Monocitos/metabolismo , Células Mieloides/inmunología , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/fisiología , Neoplasias/inmunología , Receptores CCR2/inmunología
14.
Cell Immunol ; 363: 104312, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33652258

RESUMEN

Myeloid-derived suppressor cells (MDSCs) impair protective anti-tumor immunity and remain major obstacles that stymie the effectiveness of promising cancer therapies. Diverse tumor-derived stressors galvanize the differentiation, intra-tumoral expansion, and immunomodulatory function of MDSCs. These tumor-associated 'axes of stress' underwrite the immunosuppressive programming of MDSCs in cancer and contribute to the phenotypic/functional heterogeneity that characterize tumor-MDSCs. This review discusses various tumor-associated axes of stress that direct MDSC development, accumulation, and immunosuppressive function, as well as current strategies aimed at overcoming the detrimental impact of MDSCs in cancer. To better understand the constellation of signals directing MDSC biology, we herein summarize the pivotal roles, signaling mediators, and effects of reactive oxygen/nitrogen species-related stress, chronic inflammatory stress, hypoxia-linked stress, endoplasmic reticulum stress, metabolic stress, and therapy-associated stress on MDSCs. Although therapeutic targeting of these processes remains mostly pre-clinical, intercepting signaling through the axes of stress could overcome MDSC-related immune suppression in tumor-bearing hosts.


Asunto(s)
Células Supresoras de Origen Mieloide/inmunología , Neoplasias/fisiopatología , Estrés Fisiológico/fisiología , Diferenciación Celular , Línea Celular Tumoral , Estrés del Retículo Endoplásmico/inmunología , Estrés del Retículo Endoplásmico/fisiología , Humanos , Terapia de Inmunosupresión/métodos , Células Mieloides/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/fisiología , Neoplasias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/inmunología , Estrés Fisiológico/inmunología
15.
J Immunol ; 203(5): 1095-1103, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31427398

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are immature suppressive cells found in tumors and immunological niches. In this article, we highlight the ability of MDSCs to promote IL-17-producing T cells (Th17) and regulatory T cells in addition to suppressing cytotoxic T cells in different tumor models. These interactions between MDSCs and T cells support tumor growth because IL-17 is tumorigenic in many cancer types and regulatory T cells suppress antitumor T cells. Besides T cells, MDSCs promote regulatory B cells and suppress overall B cell function; however, tumor-evoked regulatory B cells also regulate MDSC function, suggesting cross-regulation between MDSCs and B cells. These multiple functions shed light on how MDSCs dysregulate several arms of host immune response. Moreover, MDSCs promote tumor cell survival and angiogenesis to support tumors. Therefore, the multifunctional feature of MDSCs make them attractive immunotherapeutic targets.


Asunto(s)
Células Supresoras de Origen Mieloide/fisiología , Neoplasias/inmunología , Animales , Linfocitos B/inmunología , Comunicación Celular , Humanos , Inmunoterapia , Interleucina-17/biosíntesis , Neoplasias/patología , Neoplasias/terapia , Linfocitos T Reguladores/inmunología , Células Th17/inmunología
16.
J Immunol ; 202(1): 93-104, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30487174

RESUMEN

Common variable immunodeficiency disorders (CVID) represent a group of primary immunodeficiency diseases characterized by hypogammaglobulinemia and impaired specific Ab response, resulting in recurrent infections due to dysfunctional immune response. The specific mechanisms mediating immune deficiency in CVID remain to be determined. Previous studies indicated that immune dysregulation in CVID patients is associated with chronic microbial translocation, systemic immune activation, and altered homeostasis of lymphocytic and myeloid lineages. A detailed phenotypic, functional characterization of plasma markers and immune cell populations was performed in 46 CVID patients and 44 healthy donors. CVID patients displayed significantly elevated plasma levels of a marker of neutrophil activation neutrophil gelatinase-associated lipocalin. Neutrophils from CVID patients exhibited elevated surface levels of CD11b and PD-L1 and decreased levels of CD62L, CD16, and CD80, consistent with a phenotype of activated neutrophils with suppressive properties. Neutrophils from CVID patients actively suppressed T cell activation and release of IFN-γ via the production of reactive oxygen species. Furthermore, CVID was associated with an increased frequency of low-density neutrophils (LDNs)/granulocytic myeloid-derived suppressor cells. LDN/granulocytic myeloid-derived suppressor cell frequency in CVID patients correlated with reduced T cell responsiveness. Exogenous stimulation of whole blood with bacterial LPS emulated some but not all of the phenotypic changes observed on neutrophils from CVID patients and induced neutrophil population with LDN phenotype. The presented data demonstrate that neutrophils in the blood of CVID patients acquire an activated phenotype and exert potent T cell suppressive activity. Specific targeting of myeloid cell-derived suppressor activity represents a novel potential therapeutic strategy for CVID.


Asunto(s)
Inmunodeficiencia Variable Común/inmunología , Granulocitos/fisiología , Lipocalina 2/sangre , Células Supresoras de Origen Mieloide/fisiología , Neutrófilos/fisiología , Linfocitos T/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Antígeno B7-H1/metabolismo , Antígeno CD11b/metabolismo , Células Cultivadas , Femenino , Humanos , Tolerancia Inmunológica , Masculino , Persona de Mediana Edad , Activación Neutrófila , Especies Reactivas de Oxígeno/metabolismo , Adulto Joven
17.
J Am Soc Nephrol ; 31(4): 731-746, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32132198

RESUMEN

BACKGROUND: Granulocyte colony-stimulating factor (G-CSF) can increase populations of myeloid-derived suppressor cells, innate immune suppressors that play an immunoregulatory role in antitumor immunity. However, the roles of myeloid-derived suppressor cells and G-CSF in renal ischemia-reperfusion injury remain unclear. METHODS: We used mouse models of ischemia-reperfusion injury to investigate whether G-CSF can attenuate renal injury by increasing infiltration of myeloid-derived suppressor cells into kidney tissue. RESULTS: G-CSF treatment before ischemia-reperfusion injury subsequently attenuated acute renal dysfunction, tissue injury, and tubular apoptosis. Additionally, G-CSF treatment suppressed renal infiltration of macrophages and T cells as well as renal levels of IL-6, MCP-1, IL-12, TNF-α, and IFN-γ, but it increased levels of IL-10, arginase-1, and reactive oxygen species. Moreover, administering G-CSF after ischemia-reperfusion injury improved the recovery of renal function and attenuated renal fibrosis on day 28. G-CSF treatment increased renal infiltration of myeloid-derived suppressor cells (F4/80-CD11b+Gr-1int), especially the granulocytic myeloid-derived suppressor cell population (CD11b+Ly6GintLy6Clow); splenic F4/80-CD11b+Gr-1+ cells sorted from G-CSF-treated mice displayed higher levels of arginase-1, IL-10, and reactive oxygen species relative to those from control mice. Furthermore, these splenic cells effectively suppressed in vitro T cell activation mainly through arginase-1 and reactive oxygen species, and their adoptive transfer attenuated renal injury. Combined treatment with anti-Gr-1 and G-CSF showed better renoprotective effects than G-CSF alone, whereas preferential depletion of myeloid-derived suppressor cells by pep-G3 or gemcitabine abrogated the beneficial effects of G-CSF against renal injury. CONCLUSIONS: G-CSF induced renal myeloid-derived suppressor cells, thereby attenuating acute renal injury and chronic renal fibrosis after ischemia-reperfusion injury. These results suggest therapeutic potential of myeloid-derived suppressor cells and G-CSF in renal ischemia-reperfusion injury.


Asunto(s)
Lesión Renal Aguda/prevención & control , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Células Supresoras de Origen Mieloide/fisiología , Daño por Reperfusión/prevención & control , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología
18.
Br J Cancer ; 123(9): 1377-1386, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32747748

RESUMEN

BACKGROUND: BTC is an aggressive disease exacerbated by inflammation and immune suppression. Expansion of immunosuppressive cells occurs in biliary tract cancer (BTC), yet the role of BTC-derived cytokines in this process is unclear. METHODS: Activated signalling pathways and cytokine production were evaluated in a panel of human BTC cell lines. Human peripheral blood mononuclear cells (PBMCs) were cultured with BTC supernatants, with and without cytokine neutralising antibodies, and analysed by flow cytometry or immunoblot. A human BTC tissue microarray (TMA, n = 69) was stained for IL-6, GM-CSF, and CD33+S100a9+ cells and correlated with clinical outcomes. RESULTS: Immunomodulatory factors (IL-6, GM-CSF, MCP-1) were present in BTC supernatants. BTC supernatants expanded CD33dimCD11b+HLA-DRlow/- myeloid-derived suppressor cells (MDSCs) from human PBMCs. Neutralisation of IL-6 and GM-CSF in BTC supernatants inhibited activation of STAT3/5, respectively, in PBMCs, with heterogeneous effects on MDSC expansion in vitro. Staining of a BTC TMA revealed a positive correlation between IL-6 and GM-CSF, with each cytokine and more CD33+S100a9+ cells. Increased CD33+S100a9+ staining positively correlated with higher tumour grade, differentiation and the presence of satellite lesions. CONCLUSION: BTC-derived factors promote suppressive myeloid cell expansion, and higher numbers of CD33+S100a9+ cells in resectable BTC tumours correlates with more aggressive disease.


Asunto(s)
Neoplasias del Sistema Biliar/metabolismo , Neoplasias del Sistema Biliar/patología , Proliferación Celular/efectos de los fármacos , Citocinas/farmacología , Células Supresoras de Origen Mieloide/efectos de los fármacos , Calgranulina B/metabolismo , Recuento de Células , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Medios de Cultivo Condicionados/farmacología , Citocinas/metabolismo , Humanos , Activación de Linfocitos/efectos de los fármacos , Células Mieloides/efectos de los fármacos , Células Mieloides/patología , Células Mieloides/fisiología , Células Supresoras de Origen Mieloide/patología , Células Supresoras de Origen Mieloide/fisiología , Clasificación del Tumor , Invasividad Neoplásica , Lectina 3 Similar a Ig de Unión al Ácido Siálico/metabolismo
19.
Cancer Immunol Immunother ; 69(2): 215-221, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31501954

RESUMEN

Myeloid-derived suppressor cells (MDSC) are present in most cancer patients where they are significant contributors to the immune suppressive tumor microenvironment (TME). The TME is a hostile locale due to deficiencies in oxygen (hypoxia) and nutrients, and the presence of reactive oxygen species (ROS). The survival of tumor cells within the TME is partially governed by two mechanisms: (1) Activation of the transcription factor Nuclear Factor Erythroid-derived 2-like 2 (Nrf2) which turns on genes that attenuate oxidative stress; and (2) The presence of High Mobility Group Box Protein-1 (HMGB1), a damage-associated molecular pattern molecule (DAMP) that induces autophagy and protects against apoptosis. Because Nrf2 and HMGB1 promote tumor cell survival, we speculated that Nrf2 and HMGB1 may facilitate MDSC survival. We tested this hypothesis using Nrf2+/+ and Nrf2-/- BALB/c and C57BL/6 mice and pharmacological inhibitors of HMGB1. In vitro and in vivo studies demonstrated that Nrf2 increased the suppressive potency and quantity of tumor-infiltrating MDSC by up-regulating MDSC production of H2O2 and decreasing MDSC apoptosis. Decreased apoptosis was accompanied by a decrease in the production of MDSC, demonstrating that MDSC levels are homeostatically regulated. Pharmacological inhibition of autophagy increased MDSC apoptosis, indicating that autophagy increases MDSC half-life. Inhibition of HMGB1 also increased MDSC apoptosis and reduced MDSC autophagy. These results combined with our previous findings that HMGB1 drives the accumulation of MDSC demonstrate that HMGB1 maintains MDSC viability by inducing autophagy. Collectively, these findings identify Nrf2 and HMGB1 as important factors that enable MDSC to survive in the TME.


Asunto(s)
Proteína HMGB1/fisiología , Células Supresoras de Origen Mieloide/fisiología , Factor 2 Relacionado con NF-E2/fisiología , Microambiente Tumoral , Animales , Apoptosis , Autofagia , Supervivencia Celular , Humanos , Ratones , Estrés Oxidativo
20.
Hepatology ; 70(5): 1564-1581, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31021443

RESUMEN

Receptor-interacting protein kinase 3 (RIP3) is the core regulator that switches cell death from apoptosis to necrosis. However, its role in tumor immunity is unknown. In this study, decreased RIP3 expression was observed in patients with hepatocellular carcinoma (HCC), which correlates with myeloid-derived suppressor cell (MDSC) accumulation. Moreover, RIP3 is a prognosis factor for patients with HCC. We further found that RIP3 knockdown results in an increase of MDSCs and a decrease of interferon gamma-positive (IFN-γ+ ) cluster of differentiation 8-positive (CD8+ ) tumor-infiltrating lymphocytes (IFN-γ+ CD8+ T cells) in hepatoma tissues, thus promoting immune escape and HCC growth in immunocompetent mice. By phosphorylating P65Ser536 and promoting phosphorylated P65Ser536 nuclear translocation, RIP3 knockdown increases the expression of chemokine (C-X-C motif) ligand 1 (CXCL1) in HCC cells. RIP3 knockdown induces MDSC recruitment through the CXCL1-chemokine (C-X-C motif) receptor 2 (CXCR2) axis. Furthermore, a CXCR2 antagonist substantially suppresses MDSC chemotaxis and HCC growth in RIP3 knockout mice. Conclusion: RIP3 deficiency is an essential factor directing MDSC homing to HCC and promoting CXCL1/CXCR2-induced MDSC chemotaxis to facilitate HCC immune escape and HCC progression; blocking the CXCL1-CXCR2 chemokine axis may provide an immunological therapeutic approach to suppress progression of RIP3 deficiency HCC.


Asunto(s)
Carcinoma Hepatocelular/patología , Quimiocina CXCL1/fisiología , Neoplasias Hepáticas/patología , Células Supresoras de Origen Mieloide/fisiología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Receptores de Interleucina-8B/fisiología , Animales , Quimiotaxis , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA