Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 164
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
Nature ; 559(7714): 363-369, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29950727

RESUMEN

Patients with prostate cancer frequently show resistance to androgen-deprivation therapy, a condition known as castration-resistant prostate cancer (CRPC). Acquiring a better understanding of the mechanisms that control the development of CRPC remains an unmet clinical need. The well-established dependency of cancer cells on the tumour microenvironment indicates that the microenvironment might control the emergence of CRPC. Here we identify IL-23 produced by myeloid-derived suppressor cells (MDSCs) as a driver of CRPC in mice and patients with CRPC. Mechanistically, IL-23 secreted by MDSCs can activate the androgen receptor pathway in prostate tumour cells, promoting cell survival and proliferation in androgen-deprived conditions. Intra-tumour MDSC infiltration and IL-23 concentration are increased in blood and tumour samples from patients with CRPC. Antibody-mediated inactivation of IL-23 restored sensitivity to androgen-deprivation therapy in mice. Taken together, these results reveal that MDSCs promote CRPC by acting in a non-cell autonomous manner. Treatments that block IL-23 can oppose MDSC-mediated resistance to castration in prostate cancer and synergize with standard therapies.


Asunto(s)
Interleucina-23/antagonistas & inhibidores , Interleucina-23/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Neoplasias de la Próstata Resistentes a la Castración/terapia , Antagonistas de Receptores Androgénicos/farmacología , Antagonistas de Receptores Androgénicos/uso terapéutico , Andrógenos/deficiencia , Animales , Benzamidas , Proliferación Celular , Supervivencia Celular , Humanos , Interleucina-23/sangre , Interleucina-23/inmunología , Masculino , Ratones , Células Supresoras de Origen Mieloide/citología , Células Supresoras de Origen Mieloide/inmunología , Nitrilos , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Feniltiohidantoína/análogos & derivados , Feniltiohidantoína/farmacología , Feniltiohidantoína/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/sangre , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/metabolismo , Receptores de Interleucina/metabolismo , Transducción de Señal
3.
J Immunol ; 206(9): 2052-2060, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33820854

RESUMEN

RUNX1 overlapping RNA (RUNXOR) is a long noncoding RNA and a key regulator of myeloid-derived suppressor cells (MDSCs) via targeting runt-related transcription factor 1 (RUNX1). We and others have previously reported MDSC expansion and inhibition of host immune responses during viral infections; however, the mechanisms regulating MDSC differentiation and suppressive functions, especially the role of RUNXOR-RUNX1 in the regulation of MDSCs in people living with HIV (PLHIV), remain unknown. In this study, we demonstrate that RUNXOR and RUNX1 expressions are upregulated in MDSCs that expand and accumulate in human PBMCs derived from PLHIV. We found that the upregulation of RUNXOR and RUNX1 is associated with the expressions of several key immunosuppressive molecules, including arginase 1, inducible NO synthase, STAT3, IL-6, and reactive oxygen species. RUNXOR and RUNX1 could positively regulate each other's expression and control the expressions of these suppressive mediators. Specifically, silencing RUNXOR or RUNX1 expression in MDSCs from PLHIV attenuated MDSC expansion and immunosuppressive mediator expressions, whereas overexpressing RUNXOR in CD33+ myeloid precursors from healthy subjects promoted their differentiation into MDSCs and enhanced the expression of these mediators. Moreover, loss of RUNXOR-RUNX1 function in MDSCs improved IFN-γ production from cocultured autologous CD4 T cells derived from PLHIV. These results suggest that the RUNXOR-RUNX1 axis promotes the differentiation and suppressive functions of MDSCs via regulating multiple immunosuppressive signaling molecules and may represent a potential target for immunotherapy in conjunction with antiviral therapy in PLHIV.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Regulación de la Expresión Génica , Infecciones por VIH/genética , Células Supresoras de Origen Mieloide/metabolismo , ARN Largo no Codificante/genética , Arginasa/genética , Arginasa/metabolismo , Diferenciación Celular/genética , Proliferación Celular/genética , Células Cultivadas , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/metabolismo , Humanos , Células Supresoras de Origen Mieloide/citología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/genética , Regulación hacia Arriba
4.
Nature ; 543(7647): 728-732, 2017 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-28321130

RESUMEN

A significant fraction of patients with advanced prostate cancer treated with androgen deprivation therapy experience relapse with relentless progression to lethal metastatic castration-resistant prostate cancer (mCRPC). Immune checkpoint blockade using antibodies against cytotoxic-T-lymphocyte-associated protein 4 (CTLA4) or programmed cell death 1/programmed cell death 1 ligand 1 (PD1/PD-L1) generates durable therapeutic responses in a significant subset of patients across a variety of cancer types. However, mCRPC showed overwhelming de novo resistance to immune checkpoint blockade, motivating a search for targeted therapies that overcome this resistance. Myeloid-derived suppressor cells (MDSCs) are known to play important roles in tumour immune evasion. The abundance of circulating MDSCs correlates with prostate-specific antigen levels and metastasis in patients with prostate cancer. Mouse models of prostate cancer show that MDSCs (CD11b+Gr1+) promote tumour initiation and progression. These observations prompted us to hypothesize that robust immunotherapy responses in mCRPC may be elicited by the combined actions of immune checkpoint blockade agents together with targeted agents that neutralize MDSCs yet preserve T-cell function. Here we develop a novel chimaeric mouse model of mCRPC to efficiently test combination therapies in an autochthonous setting. Combination of anti-CTLA4 and anti-PD1 engendered only modest efficacy. Targeted therapy against mCRPC-infiltrating MDSCs, using multikinase inhibitors such as cabozantinib and BEZ235, also showed minimal anti-tumour activities. Strikingly, primary and metastatic CRPC showed robust synergistic responses when immune checkpoint blockade was combined with MDSC-targeted therapy. Mechanistically, combination therapy efficacy stemmed from the upregulation of interleukin-1 receptor antagonist and suppression of MDSC-promoting cytokines secreted by prostate cancer cells. These observations illuminate a clinical path hypothesis for combining immune checkpoint blockade with MDSC-targeted therapies in the treatment of mCRPC.


Asunto(s)
Inmunoterapia/métodos , Neoplasias de la Próstata Resistentes a la Castración/inmunología , Neoplasias de la Próstata Resistentes a la Castración/terapia , Anilidas/farmacología , Anilidas/uso terapéutico , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Quimera , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Humanos , Imidazoles/farmacología , Imidazoles/uso terapéutico , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Ratones , Terapia Molecular Dirigida , Células Supresoras de Origen Mieloide/citología , Células Supresoras de Origen Mieloide/efectos de los fármacos , Células Supresoras de Origen Mieloide/inmunología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Neoplasias de la Próstata Resistentes a la Castración/patología , Piridinas/farmacología , Piridinas/uso terapéutico , Quinolinas/farmacología , Quinolinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
5.
Cell Mol Life Sci ; 78(23): 7161-7183, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34635950

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells (IMCs) with immunosuppressive functions, whereas IMCs originally differentiate into granulocytes, macrophages, and dendritic cells (DCs) to participate in innate immunity under steady-state conditions. At present, difficulties remain in identifying MDSCs due to lacking of specific biomarkers. To make identification of MDSCs accurately, it also needs to be determined whether having immunosuppressive functions. MDSCs play crucial roles in anti-tumor, angiogenesis, and metastasis. Meanwhile, MDSCs could make close interaction with osteoclasts, osteoblasts, chondrocytes, and other stromal cells within microenvironment of bone and joint, and thereby contributing to poor prognosis of bone-related diseases such as cancer-related bone metastasis, osteosarcoma (OS), rheumatoid arthritis (RA), osteoarthritis (OA), and orthopedic trauma. In addition, MDSCs have been shown to participate in the procedure of bone repair. In this review, we have summarized the function of MDSCs in cancer-related bone metastasis, the interaction with stromal cells within the bone microenvironment as well as joint microenvironment, and the critical role of MDSCs in bone repair. Besides, the promising value of MDSCs in the treatment for bone-related diseases is also well discussed.


Asunto(s)
Neoplasias Óseas/patología , Regeneración Ósea/fisiología , Tolerancia Inmunológica/inmunología , Células Supresoras de Origen Mieloide/citología , Artritis Reumatoide/patología , Enfermedades Óseas/patología , Neoplasias Óseas/secundario , Microambiente Celular/fisiología , Células Dendríticas/citología , Granulocitos/citología , Humanos , Inmunidad Innata/inmunología , Macrófagos/citología , Células Supresoras de Origen Mieloide/inmunología , Osteoartritis/patología
6.
Cell Mol Life Sci ; 78(1): 173-193, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32654036

RESUMEN

The successful treatment of human cancers by immunotherapy has been made possible by breakthroughs in the discovery of immune checkpoint regulators, including CTLA-4 and PD-1/PD-L1. However, the immunosuppressive effect of the tumor microenvironment still represents an important bottleneck that limits the success of immunotherapeutic approaches. The tumor microenvironment influences the metabolic crosstalk between tumor cells and tumor-infiltrating immune cells, creating competition for the utilization of nutrients and promoting immunosuppression. In addition, tumor-derived metabolites regulate the activation and effector function of immune cells through a variety of mechanisms; in turn, the metabolites and other factors secreted by immune cells can also become accomplices to cancer development. Immune-metabolic checkpoint regulation is an emerging concept that is being studied with the aim of restoring the immune response in the tumor microenvironment. In this review, we summarize the metabolic reprogramming of various cell types present in the tumor microenvironment, with a focus on the interaction between the metabolic pathways of these cells and antitumor immunosuppression. We also discuss the main metabolic checkpoints that could provide new means of enhancing antitumor immunotherapy.


Asunto(s)
Inmunoterapia , Neoplasias/patología , Microambiente Tumoral , Antígeno B7-H1/inmunología , Antígeno B7-H1/metabolismo , Antígeno CTLA-4/inmunología , Antígeno CTLA-4/metabolismo , Humanos , Células Supresoras de Origen Mieloide/citología , Células Supresoras de Origen Mieloide/metabolismo , Neoplasias/inmunología , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Macrófagos Asociados a Tumores/citología , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo
7.
Am J Physiol Cell Physiol ; 321(3): C569-C584, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34288720

RESUMEN

Rheumatoid arthritis (RA) is a debilitating autoimmune disease of unknown cause, characterized by infiltration and accumulation of activated immune cells in the synovial joints where cartilage and bone destructions occur. Myeloid-derived suppressor cells (MDSCs) are of myeloid origin and are able to suppress T cell responses. Src homology 2 domain-containing inositol polyphosphate 5-phosphatase 1 (SHIP1) was shown to be involved in the regulation of MDSC differentiation. The purpose of the present study was to investigate the effect of inhibition of SHIP1 on the expansion of MDSCs in RA using a collagen-induced inflammatory arthritis (CIA) mouse model. In DBA/1 mice, treatment with a small molecule-specific SHIP1 inhibitor 3α-aminocholestane (3AC) induced a marked expansion of MDSCs in vivo. Both pretreatment with 3AC of DBA/1 mice prior to CIA induction and intervention with 3AC during CIA progression significantly reduced disease incidence and severity. Adoptive transfer of MDSCs isolated from 3AC-treated mice, but not naïve MDSCs from normal mice, into CIA mice significantly reduced disease incidence and severity, indicating that the 3AC-induced MDSCs were the cellular mediators of the observed amelioration of the disease. In conclusion, inhibition of SHIP1 expands MDSCs in vivo and attenuates development of CIA in mice. Small molecule-specific inhibition of SHIP1 may therefore offer therapeutic benefit to patients with RA and other autoimmune diseases.


Asunto(s)
Antiinflamatorios/farmacología , Artritis Experimental/tratamiento farmacológico , Colestanos/farmacología , Células Supresoras de Origen Mieloide/inmunología , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/genética , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Artritis Experimental/genética , Artritis Experimental/inmunología , Artritis Experimental/patología , Artritis Reumatoide/genética , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Comunicación Celular , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Expresión Génica , Humanos , Cápsula Articular/efectos de los fármacos , Cápsula Articular/inmunología , Cápsula Articular/patología , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Células Supresoras de Origen Mieloide/citología , Células Supresoras de Origen Mieloide/trasplante , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/antagonistas & inhibidores , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/inmunología , Índice de Severidad de la Enfermedad , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/patología
8.
Infect Immun ; 89(11): e0040721, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34370509

RESUMEN

During chronic infection with Helicobacter pylori, Schlafen 4-expressing myeloid-derived suppressor cells (SLFN4+ MDSCs) create a microenvironment favoring intestinal metaplasia and neoplastic transformation. SLFN4 can be induced by alpha interferon (IFN-α), which is mainly secreted from plasmacytoid dendritic cells (pDCs). This study tested the hypothesis that Helicobacter pylori infection promotes SLFN4+ MDSC differentiation by inducing pDCs to secrete IFN-α. C57BL/6 mice were gavaged with H. pylori, and infection lasted 2, 4, or 6 months. Mouse pDCs were isolated from bone marrow of wild-type C57BL/6J mice. The results showed that H. pylori infection increased the number of SLFN4+ MDSCs by inducing IFN-α expression in mice. Further mechanistic experiments unraveled that IFN-α induced SLFN4 transcription by binding to the Slfn4 promoter. Furthermore, H. pylori infection stimulated pDCs to secrete IFN-α by activating the TLR9-MyD88-IRF7 pathway. Collectively, Helicobacter pylori infection promotes SLFN4+ MDSC differentiation by inducing secretion of IFN-α from pDCs.


Asunto(s)
Proteínas Portadoras/genética , Células Dendríticas/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter pylori , Interferón Tipo I/biosíntesis , Células Supresoras de Origen Mieloide/citología , Animales , Diferenciación Celular , Factor 7 Regulador del Interferón/fisiología , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/fisiología , Regiones Promotoras Genéticas , Receptor Toll-Like 9/fisiología
9.
Cancer Sci ; 112(11): 4470-4477, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34464993

RESUMEN

Fusobacterium nucleatum has been detected in 8%-13% of human colorectal cancer, and shown to inhibit immune responses against primary colorectal tumors in animal models. Thus, we hypothesized that the presence of F. nucleatum might be associated with reduced T cell density in colorectal cancer liver metastases (CRLM). We quantified F. nucleatum DNA in 181 CRLM specimens using quantitative PCR assay. The densities of CD8+ T cells, CD33+ cells (marker for myeloid-derived suppressor cells [MDSCs]), and CD163+ cells (marker for tumor-associated macrophages [TAMs]) in CRLM tissue were determined by immunohistochemical staining. Fusobacterium nucleatum was detected in eight (4.4%) of 181 CRLM specimens. Compared with F. nucleatum-negative CRLM, F. nucleatum-positive CRLM showed significantly lower density of CD8+ T cells (P = .033) and higher density of MDSCs (P = .001). The association of F. nucleatum with the density of TAMs was not statistically significant (P = .70). The presence of F. nucleatum is associated with a lower density of CD8+ T cells and a higher density of MDSCs in CRLM tissue. Upon validation, our findings could provide insights to develop strategies that involve targeting microbiota and immune cells for the prevention and treatment of CRLM.


Asunto(s)
Linfocitos T CD8-positivos/citología , Neoplasias Colorrectales/microbiología , Fusobacterium nucleatum/inmunología , Neoplasias Hepáticas/inmunología , Neoplasias Colorrectales/patología , ADN Bacteriano/análisis , Femenino , Fusobacterium nucleatum/genética , Fusobacterium nucleatum/aislamiento & purificación , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/microbiología , Neoplasias Hepáticas/secundario , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Células Supresoras de Origen Mieloide/citología , Macrófagos Asociados a Tumores/citología
10.
Cell Immunol ; 359: 104258, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33338939

RESUMEN

Collaborative research is reviewed in which mass spectrometry-based proteomics and next generation sequencing were used qualitatively and quantitatively to interrogate proteins and RNAs carried in intact myeloid-derived suppressor cells (MDSC) and exosomes shed in vitro by MDSC. In aggregate exosomes more than 4000 proteins were identified, including annexins and immunosuppressive mediators. Bioassays showed that exosomes induce MDSC chemotaxis dependent on S100A8 and S100A9 in their cargo. Surface selective chemistry identified glycoproteins on MDSC and exosome surfaces, including CD47 and thrombospondin 1, which both facilitate exosome-catalyzed chemotaxis. Large numbers of mRNAs and microRNAs were identified in aggregate exosomes, whose potential functions in receptor cells include angiogenesis, and proinflammatory and immunosuppressive activities. Inflammation was found to have asymmetric effects on MDSC and exosomal cargos. Collectively, our findings indicate that the exosomes shed by MDSC provide divergent and complementary functions that support the immunosuppression and tumor promotion activities of MDSC.


Asunto(s)
Exosomas/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Transporte de Proteínas/fisiología , Línea Celular Tumoral , Exosomas/fisiología , Humanos , Inflamación/patología , MicroARNs/metabolismo , Células Supresoras de Origen Mieloide/citología , Proteínas/metabolismo , ARN Mensajero/metabolismo
11.
Cell Immunol ; 363: 104317, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33714729

RESUMEN

Myeloid derived suppressor cells (MDSCs) can be subset into monocytic (M-), granulocytic (G-) or polymorphonuclear (PMN-), and immature (i-) or early MDSCs and have a role in many disease states. In cancer patients, the frequencies of MDSCs can positively correlate with stage, grade, and survival. Most clinical studies into MDSCs have been undertaken with peripheral blood (PB); however, in the present studies, we uniquely examined MDSCs in the spleens and PB from patients with gastrointestinal cancers. In our studies, MDSCs were rigorously subset using the following markers: Lineage (LIN) (CD3, CD19 and CD56), human leukocyte antigen (HLA)-DR, CD11b, CD14, CD15, CD33, CD34, CD45, and CD16. We observed a significantly higher frequency of PMN- and M-MDSCs in the PB of cancer patients as compared to their spleens. Expression of the T-cell suppressive enzymes arginase (ARG1) and inducible nitric oxide synthase (i-NOS) were higher on all MDSC subsets for both cancer patients PB and spleen cells as compared to MDSCs from the PB of normal donors. Similar findings for the activation markers lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1), program death ligand 1 (PD-L1) and program cell death protein 1 (PD-1) were observed. Interestingly, the total MDSC cell number exported to clustering analyses was similar between all sample types; however, clustering analyses of these MDSCs, using these markers, uniquely documented novel subsets of PMN-, M- and i-MDSCs. In summary, we report a comparison of splenic MDSC frequency, subtypes, and functionality in cancer patients to their PB by clustering and cytometric analyses.


Asunto(s)
Células Supresoras de Origen Mieloide/metabolismo , Bazo/inmunología , Adulto , Anciano , Arginasa/metabolismo , Antígeno B7-H1/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Análisis por Conglomerados , Femenino , Citometría de Flujo/métodos , Neoplasias Gastrointestinales/inmunología , Antígenos HLA-DR/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Células Supresoras de Origen Mieloide/citología , Células Supresoras de Origen Mieloide/inmunología , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Receptores Depuradores de Clase E/metabolismo , Bazo/patología
12.
Exp Cell Res ; 394(2): 112119, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32485182

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are a significant obstacle for immunotherapy of cancer. It is of great clinical relevance to study the mechanism of MDSCs accumulation in mouse spleens and establish a stable method to obtain high-purity MDSCs in vitro for further research. Here, we established a new method for amplifying a large number of highly pure MDSCs in vitro. To mimic the microenvironment of MDSCs development in vivo, mouse splenic stroma feeder cells and serum-free medium containing granulocyte-macrophage colony stimulating factor (GM-CSF) were used to induce myeloid precursors in mouse bone marrow cells, which differentiate into MDSCs. Development and immunological functions of the cells were monitored both in vivo and in vitro. A total of 4 × 108 MDSCs could be obtained from the bone marrow from one mouse, the ratio of CD11b+Gr-1+ MDSCs could reach 93.8% ± 3.3% after nine days of culture in vitro. Cultured MDSCs maintained a similar immunophenotype with MDSCs found in tumor-bearing mice. Colony forming assay in vitro and in vivo demonstrated that these were myeloid precursor cells. These cells generated high levels of reactive oxygen species and arginase 1 to prevent proliferation of CD8+ T cells in vitro. These also increased regulatory T (Treg) cells in blood while promoting the growth of lymphoma in vivo. In addition, cultured MDSCs effectively inhibited acute graft-versus-host disease (aGVHD). Our findings suggest that mouse splenic stroma plays an important role in the generation of MDSCs and represent a preliminary mechanism for the accumulation of MDSCs in spleens, and thereby lay the foundation for basic research and the clinical application of MDSCs.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Nutrientes/citología , Células Supresoras de Origen Mieloide/citología , Bazo/citología , Animales , Arginasa/metabolismo , Linfocitos T CD8-positivos/citología , Proliferación Celular , Ensayo de Unidades Formadoras de Colonias , Femenino , Enfermedad Injerto contra Huésped/inmunología , Inmunofenotipificación , Linfoma/patología , Masculino , Ratones , Especies Reactivas de Oxígeno/metabolismo , Células del Estroma/citología , Análisis de Supervivencia , Linfocitos T Reguladores/citología
13.
Prep Biochem Biotechnol ; 51(5): 415-421, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32985918

RESUMEN

Spleen is an information-rich and easy-accessible peripheral lymphoid organ. It has complex cell composition because of the immunocytes maturity and settle down. Changes of the composition and function of these immunocytes are critical to body immune response. To understand the cell behaviors, specific cell subpopulations are required to be separated without heterogeneity. Density gradient centrifugation is one of the cell separation methods with high throughput. However, the greatest defect of this method is its low cell purity. In this study, the separation conditions of tumor-bearing mouse splenocytes were optimized by separation solutions with different density gradients. After separation, lymphocytes were located at the second layer with the proportion of 84.9%, monocytic-like myeloid-derived suppressor cells (Mo-MDSCs) were located at the fourth layer with the proportion of 54.2% and polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) were located at the sixth layer with the proportion of 85.5%. Cells in different layers were further determined by verifying the gene expression pattern of some chemokine receptors on cell surfaces. Furthermore, this method was also used to separate healthy mouse splenocytes. Therefore, this method will be highly useful to separate mouse splenocytes and has laid a foundation for further research on the changes and roles of immunocytes during the development of cancer.


Asunto(s)
Separación Celular , Células Supresoras de Origen Mieloide/citología , Bazo/citología , Animales , Línea Celular Tumoral , Centrifugación por Gradiente de Densidad , Femenino , Ratones , Ratones Endogámicos BALB C
14.
Inflammopharmacology ; 29(6): 1613-1624, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34613567

RESUMEN

The most fatal malignancy of the central nervous system (CNS) is glioblastoma. Brain cancer is a 'cold' tumor because of fewer immunoregulatory cells and more immunosuppressive cells. Due to the cold nature of brain cancers, conventional treatments which are used to manage glioma patients show little effectiveness. Glioma patients even showed resistance to immune checkpoint blockade (ICB) and no significant efficacy. It has been shown that myeloid-derived suppressor cells (MDSCs) account for approximately 30-50% of the tumor mass in glioma. This study aimed to review MDSC function in brain cancer, as well as possible treatments and related challenges. In brain cancer and glioma, several differences in the context of MDSCs have been reported, including disagreements about the MDSC subtype that has the most inhibitory function in the brain, or inhibitory function of regulatory B cells (Bregs). There are also serious challenges in treating glioma patients. In addition to the cold nature of glioma, there are reports of an increase in MDSCs following conventional chemotherapy treatments. As a result, targeting MDSCs in combination with other therapies, such as ICB, is essential, and recent studies with the combination therapy approach have shown promising therapeutic effects in brain cancer.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Células Supresoras de Origen Mieloide/citología , Animales , Antineoplásicos/farmacología , Neoplasias Encefálicas/terapia , Resistencia a Antineoplásicos , Glioblastoma/patología , Glioblastoma/terapia , Glioma/terapia , Humanos , Terapia Molecular Dirigida
15.
Cytotherapy ; 22(12): 718-733, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32811747

RESUMEN

BACKGROUND AIMS: Mesenchymal stem cells (MSCs) use multiple mechanisms to constrain both innate and adaptive immune responses to prevent graft-versus-host disease (GVHD). Myeloid-derived suppressor cells (MDSCs), as a heterogeneous population of early myeloid progenitor cells originating from bone marrow, are a naturally occurring immune regulatory population associated with inhibition of ongoing inflammatory responses, indicating their potential for GVHD therapy. There is accumulating evidence that MSCs and MDSCs do not act independently, but rather establish crosstalk. However, the role of MSCs in MDSC expansion and activation in GVHD remains unexplored. METHODS: In vitro experiments included 2 groups: peripheral blood mononuclear cells (PBMCs) after mobilization and human umbilical cord blood-derived MSCs (UCB-MSCs) co-cultured with PBMCs. The number and functional difference of MDSCs in PBMCs were determined by flow cytometry. The culture supernatants of co-cultured cells were analyzed to identify cytokines involved in MDSC proliferation. The relationship between MSCs and MDSCs was clarified in GVHD and graft-versus-leukemia (GVL) animal models. RESULTS: In vitro experiments confirmed that UCB-MSCs secreted HLA-G protein to promote and maintain the proliferation of MDSCs in peripheral blood after granulocyte colony-stimulating factor mobilization, and UCB-MSCs mediated the function of MDSCs to inhibit the proliferation of T cells and promote the proliferation of regulatory T cells. UCB-MSCs overexpressing HLA-G induced MDSC production in recipient mice, improved the ability of MDSCs to suppress T cells and further reduced acute GVHD (aGVHD) symptoms and survival time without influencing GVL effects. CONCLUSIONS: UCB-MSCs expanded MDSCs via HLA-G/Ig-like transcript 4, reducing the severity of aGVHD without affecting GVL. The immunosuppressive potential of MSCs for the treatment of aGVHD significantly affects the development of MDSCs, thereby consolidating the position of MSCs in the prevention and treatment of aGVHD.


Asunto(s)
Sangre Fetal/citología , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/prevención & control , Antígenos HLA-G/metabolismo , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Células Madre Mesenquimatosas/citología , Células Supresoras de Origen Mieloide/citología , Animales , Proliferación Celular , Enfermedad Injerto contra Huésped/inmunología , Humanos , Inmunofenotipificación , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos C57BL , Células Supresoras de Origen Mieloide/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Inmunológicos/metabolismo , Análisis de Supervivencia , Linfocitos T Reguladores/inmunología
16.
FASEB J ; 33(5): 5967-5978, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30730772

RESUMEN

Liver regeneration depends on sequential activation of pathways and cells involving the remaining organ in recovery of mass. Proliferation of parenchyma is dependent on angiogenesis. Understanding liver regeneration-associated neovascularization may be useful for development of clinical interventions. Myeloid-derived suppressor cells (MDSCs) promote tumor angiogenesis and play a role in developmental processes that necessitate rapid vascularization. We therefore hypothesized that the MDSCs could play a role in liver regeneration. Following partial hepatectomy, MDSCs were enriched within regenerating livers, and their depletion led to increased liver injury and postoperative mortality, reduced liver weights, decreased hepatic vascularization, reduced hepatocyte hypertrophy and proliferation, and aberrant liver function. Gene expression profiling of regenerating liver-derived MDSCs demonstrated a large-scale transcriptional response involving key pathways related to angiogenesis. Functionally, enhanced reactive oxygen species production and angiogenic capacities of regenerating liver-derived MDSCs were confirmed. A comparative analysis revealed that the transcriptional response of MDSCs during liver regeneration resembled that of peripheral blood MDSCs during progression of abdominal tumors, suggesting a common MDSC gene expression profile promoting angiogenesis. In summary, our study shows that MDSCs contribute to early stages of liver regeneration possibly by exerting proangiogenic functions using a unique transcriptional program.-Nachmany, I., Bogoch, Y., Sivan, A., Amar, O., Bondar, E., Zohar, N., Yakubovsky, O., Fainaru, O., Klausner, J. M., Pencovich, N. CD11b+Ly6G+ myeloid-derived suppressor cells promote liver regeneration in a murine model of major hepatectomy.


Asunto(s)
Hepatectomía , Regeneración Hepática , Células Supresoras de Origen Mieloide/citología , Animales , Antígenos Ly/metabolismo , Antígeno CD11b/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Hígado/cirugía , Masculino , Ratones , Ratones Endogámicos BALB C , Células Mieloides/citología , Neovascularización Patológica , Especies Reactivas de Oxígeno/metabolismo
17.
Cell Mol Biol (Noisy-le-grand) ; 66(2): 36-40, 2020 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-32415924

RESUMEN

Hepatocellular carcinoma is known to be a common predominant cancer in adults, especially in eastern countries. Immune response and cancer-associated fibroblasts (CAFs) have significant influences on tumor development. However, the interaction between CAFs and immunotherapy is unclear in hepatocellular carcinoma. We measured the number of activated fibroblasts in hepatocellular carcinoma samples and samples taken from normal liver tissues. A total of 20 patients' fresh hepatocellular carcinoma and normal tissues which were surrounding the tumor were obtained from the surgery and used for evaluating alpha-SMA expression. We investigated the effects of CAFs in anti-tumor immunity in hepatocellular carcinoma animal model. The effects of CAFs in inducing anti-PD-1 treatment resistance were also measured in a preclinical animal model. Activated fibroblasts were highly accumulated in hepatocellular carcinoma tissues but not in surrounding normal tissues. CAFs showed a significant tumor-promoting effect in an immunocompetent model. The infiltration and function of some immune cells like myeloid-derived suppressive cells and T-cells were increased by CAFs. CAFs also reduced the number and activation of tumor-infiltrating cytotoxic T-cell in tumor tissue. In the treatment model, tumors with a higher amount of CAFs had been insensitive to therapy with anti-PD-1. CAFs are potent inducers of immunosuppression in hepatocellular carcinoma. Depleting CAFs rescued the antitumor immunity in the hepatocellular model and could be a novel treatment to combine with the existing immunotherapy.


Asunto(s)
Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Actinas/metabolismo , Animales , Antineoplásicos/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Fibroblastos Asociados al Cáncer/citología , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/trasplante , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Citocinas/metabolismo , Dasatinib/uso terapéutico , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Femenino , Humanos , Inmunoterapia , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos BALB C , Células Supresoras de Origen Mieloide/citología , Células Supresoras de Origen Mieloide/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Trasplante Homólogo
18.
Nanomedicine ; 24: 102150, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31931230

RESUMEN

Myeloid-derived suppressor cells (MDSCs) play a pivotal role in cancer. To overcome the problem of the MDSCs in the tumor microenvironment in this study, a combination of immunotherapy and chemotherapy was used. For this purpose, a liposomal formulation of P5 peptide and PEGylated liposomal doxorubicin (Doxil®) was utilized to treat mice bearing HER2+ tumor model. The results revealed that Doxil® administration before immunotherapy had not only reduced the population and functions of the MDSCs in the spleen (P < 0.001) and the tumor microenvironment (P < 0.05) but had also supported further immunotherapy including enhanced CD4+ (P < 0.01) and CD8+ lymphocyte (P < 0.001) population and IFN-γ production (P < 0.001). This effect was also more pronounced with a liposomal P5 and Doxil® compared with free peptide and doxorubicin. In conclusion, the results demonstrated that Doxil® plus liposomal P5 could have a decreasing effect on MDSCs and tumor growth, and it could be beneficial in breast cancer treatment.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/análogos & derivados , Células Supresoras de Origen Mieloide/citología , Células Supresoras de Origen Mieloide/metabolismo , Animales , ADN Complementario/química , Doxorrubicina/química , Femenino , Citometría de Flujo , Inmunoterapia , Ratones , Ratones Endogámicos BALB C , Polietilenglicoles/química
19.
Pol J Pathol ; 71(1): 46-54, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32429654

RESUMEN

Prostate cancer is one of the most frequent cancers in men. Although several treatment options exist, their clinical effectiveness is still not satisfactory. One the possible reason of such situation might be the presence of myeloid-derived suppressor cells (MDSC) and their pro-tumorigenic activity. MDSC possess immunosuppressive ability and in many studies were shown to support tumor development and progression. In this study we addressed the question whether commonly used therapies of prostate cancer affect the level of MDSC populations in the patients' blood. We compared the level of granulocytic (Gr-MDSC), monocytic (Mo-MDSC) and early stage MDSC (eMDSC) in the blood of patients at different clinical stage and different tumor grading scores, who underwent either surgery or hormonal therapy alone or were given a combined treatment, including e.g. radiotherapy. The obtained results showed that the level of Gr-MDSC was significantly lower in all treated patients comparing to untreated group. On the other hand, surgery or hormonal therapy alone did not affect the level of Mo-MDSC. These results were independent of the PSA level, the tumor grading and clinical stage of the patients. In conclusion, we suggest that Mo-MDSC should be considered as a potential therapy target in the course of prostate cancer treatment to enhance its anti-tumor effectiveness.


Asunto(s)
Células Supresoras de Origen Mieloide/citología , Neoplasias de la Próstata/sangre , Granulocitos/citología , Humanos , Masculino , Monocitos/citología , Neoplasias de la Próstata/terapia , Resultado del Tratamiento
20.
Int J Mol Sci ; 21(10)2020 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-32443699

RESUMEN

There has been a great interest in myeloid-derived suppressor cells (MDSCs) due to their biological functions in tumor-mediated immune escape by suppressing antitumor immune responses. These cells arise from altered myelopoiesis in response to the tumor-derived factors. The most recognized function of MDSCs is suppressing anti-tumor immune responses by impairing T cell functions, and these cells are the most important players in cancer dissemination and metastasis. Therefore, understanding the factors and the mechanism of MDSC differentiation, expansion, and recruitment into the tumor microenvironment can lead to its control. However, most of the studies only defined MDSCs with no further characterization of granulocytic and monocytic subsets. In this review, we discuss the mechanisms by which specific MDSC subsets contribute to cancers. A better understanding of MDSC subset development and the specific molecular mechanism is needed to identify treatment targets. The understanding of the specific molecular mechanisms responsible for MDSC accumulation would enable more precise therapeutic targeting of these cells.


Asunto(s)
Células Supresoras de Origen Mieloide/inmunología , Mielopoyesis , Neoplasias/sangre , Animales , Humanos , Células Supresoras de Origen Mieloide/citología , Neoplasias/inmunología , Neoplasias/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA