Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros











Tipo de estudio
Intervalo de año de publicación
1.
J Leukoc Biol ; 115(5): 958-984, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38236200

RESUMEN

Myeloid-derived suppressor cells are heterogenous immature myeloid lineage cells that can differentiate into neutrophils, monocytes, and dendritic cells as well. These cells have been characterized to have potent immunosuppressive capacity in neoplasia and a neoplastic chronic inflammatory microenvironment. Increased accumulation of myeloid-derived suppressor cells was reported with poor clinical outcomes in patients. They support neoplastic progression by abrogating antitumor immunity through inhibition of lymphocyte functions and directly by facilitating tumor development. Yet the shifting genetic signatures of this myeloid lineage cell toward immunosuppressive functionality in progressive tumor development remain elusive. We have attempted to identify the gene expression profile using lineage-specific markers of these unique myeloid lineage cells in a tumor microenvironment and bone marrow using a liquid transplantable mice tumor model to trace the changing influence of the tumor microenvironment on myeloid-derived suppressor cells. We analyzed the phenotype, functional shift, suppressive activity, differentiation status, and microarray-based gene expression profile of CD11b+Gr1+ lineage-specific cells isolated from the tumor microenvironment and bone marrow of 4 stages of tumor-bearing mice and compared them with control counterparts. Our analysis of differentially expressed genes of myeloid-derived suppressor cells isolated from bone marrow and the tumor microenvironment reveals unique gene expression patterns in the bone marrow and tumor microenvironment-derived myeloid-derived suppressor cells. It also suggests T-cell suppressive activity of myeloid-derived suppressor cells progressively increases toward the mid-to-late phase of the tumor and a significant differentiation bias of tumor site myeloid-derived suppressor cells toward macrophages, even in the presence of differentiating agents, indicating potential molecular characteristics of myeloid-derived suppressor cells in different stages of the tumor that can emerge as an intervention target.


Asunto(s)
Diferenciación Celular , Progresión de la Enfermedad , Células Supresoras de Origen Mieloide , Microambiente Tumoral , Animales , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/patología , Microambiente Tumoral/inmunología , Ratones , Regulación Neoplásica de la Expresión Génica , Perfilación de la Expresión Génica , Ratones Endogámicos C57BL , Células de la Médula Ósea/metabolismo , Antígeno CD11b/metabolismo , Antígeno CD11b/genética , Médula Ósea/patología , Médula Ósea/metabolismo
2.
iScience ; 23(10): 101623, 2020 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-33089111

RESUMEN

Galunisertib (LY2157299) is a selective ATP-mimetic inhibitor of TGF-ß receptor-I activation, currently under clinical trial in a variety of cancers. We have tested the combined effects of galunisertib- and interleukin-15-activated dendritic cells in an aggressive and highly metastatic murine lymphoma. Based on the tumor-draining lymph node architecture, and its histology, the combination therapy results in better prognosis, including disappearance of the disease-exacerbating regulatory T cells. Our data suggest that galunisertib significantly enhances the success of immunotherapy with IL-15-activated dendritic cells by limiting the regulatory T cells generation with consequent downregulation of regulatory T cells in the tumor-draining lymph nodes and vascularized organ like spleen. This is also associated with consistent loss p-SMAD2 and downregulation of Neuropilin-1, leading to better prognosis and positive outcome. These results connect the role of combined therapy with the consequent elimination of disease-exacerbating T regulatory cells in a metastatic murine lymphoma.

3.
Life Sci ; 253: 117731, 2020 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-32353431

RESUMEN

BACKGROUND: Very little is known about the role inflammation and mechanism(s) that enables the tumor to evade host's anti-tumor immune function during very initial days of tumor establishment. Our study focuses on the immune response and local inflammation specially the pro-inflammatory and immune modifier components that are responsible for tumor-induced immune-suppression, tumor-associated macrophages (TAM) at tumor microenvironment in mouse model from very early to late phase of tumor progression. METHODS: 1 × 105 Ascites tumor, EAC in Swiss albino or Sarcoma-180 (S-180) in Balb c mice strain were inoculated intra-peritonially and grouped into Control (0 day or no tumor), initial phase (3 day tumor), early (7 Day), Late (14 day) and terminal (21 day tumor) sets. T cell activity, tumor niche macrophage, inflammatory signatures were studied using Confocal microscopy, flowcytometry, ELISA, q-RT PCR and Western blot. RESULTS: We observed increased T cell infiltration at a very early stage of tumorigenesis in the tumor site with elevated percentage of activated/memory T cells. But increased cellular death and functional suppression of tumor site T cells during final stages. We observed increased infiltration of TAMs with skewed M2 phenotype. Increased chemokine receptor expression could be noted on these TAMs. Using HIF-1α inhibitor and prostaglandin receptor antagonists we demonstrated crucial role of these factor in functional alteration in TAMs. HIF-1α inhibition and also by prostaglandin receptor inhibition reduced signature pro-inflammatory gene expression, migration of macrophages and T cell suppression capacity of TAMs. We also demonstrated that PGE2 can induce HIF-1α activation in relatively less hypoxic microenvironment during early stages of tumor. CONCLUSION: Altogether, these findings strongly suggest link between prostaglandin mediated early HIF-1α activation and subsequent hypoxia induced HIF-1α activation that further enhances prostaglandin synthesis driving the recruitment and functional alteration of tumor site macrophages.


Asunto(s)
Carcinoma de Ehrlich/patología , Dinoprostona/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/patología , Sarcoma 180/patología , Animales , Carcinoma de Ehrlich/inmunología , Movimiento Celular , Progresión de la Enfermedad , Inflamación/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Fenotipo , Sarcoma 180/inmunología , Linfocitos T/inmunología , Microambiente Tumoral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA