Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Cancers (Basel) ; 14(14)2022 Jul 19.
Article in English | MEDLINE | ID: mdl-35884574

ABSTRACT

Pro-inflammatory cytokines play key roles in elevating cancer progression in triple-negative breast cancer (TNBC). We demonstrate that specific combinations between TNFα, IL-1ß and IFNγ up-regulated the proportion of human TNBC cells co-expressing the inhibitory immune checkpoints PD-L1 and PD-L2: TNFα + IL-1ß in MDA-MB-231 cells and IFNγ + IL-1ß in BT-549 cells; in the latter cells, the process depended entirely on STAT1 activation, with no involvement of p65 (CRISPR-Cas9 experiments). Highly significant associations between the pro-inflammatory cytokines and PD-L1/PD-L2 expression were revealed in the TCGA dataset of basal-like breast cancer patients. In parallel, we found that the pro-inflammatory cytokines regulated the expression of the soluble receptors of tumor necrosis factor α (TNFα), namely sTNFR1 and sTNFR2; moreover, we revealed that sTNFR1 and sTNFR2 serve as anti-metastatic and protective factors in TNBC, reducing the TNFα-induced production of inflammatory pro-metastatic chemokines (CXCL8, CXCL1, CCL5) by TNBC cells. Importantly, we found that in the context of inflammatory stimulation and also without exposure to pro-inflammatory cytokines, elevated levels of PD-L1 have down-regulated the production of anti-tumor sTNFR1 and sTNFR2. These findings suggest that in addition to its immune-suppressive activities, PD-L1 may promote disease course in TNBC by inhibiting the protective effects of sTNFR1 and sTNFR2.

2.
Cancer Immunol Immunother ; 69(7): 1315-1326, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32198536

ABSTRACT

In view of the relatively limited efficacy of immunotherapies targeting the PD-1-PD-L1 axis in triple-negative breast cancer (TNBC) and of published reports on tumor-promoting roles of TNFR2+ tumor-infiltrating lymphocytes (TNFR2+ TILs), we determined the incidence of TNFR2+ TILs in TNBC patient tumors, their association with disease outcome and relations with PD-1+ TILs. Using a cohort of treatment-naïve TNBC patients with long follow-up (n = 70), we determined the presence of TNFR2+ TILs and PD-1+ TILs by immunohistochemistry. TILs (≥ 1% of cellular mass) and TNFR2+ TILs (≥ 1% of total TILs) were detected in 96% and 74% of tumors, respectively. The presence of TILs at > 5% of tumor cell mass ("Positive TILs"), as well as of positive TNFR2+ TILs (> 5%), was independently associated with good prognosis, and combination of both parameters demonstrated superior outcome relative to their lower levels. PD1+ TILs (> 5/hot spot) were detected in 63% of patients. High levels of PD-1+ TILs (> 20/hot spot) showed an unfavorable disease outcome, and in their presence, the favorable outcome of positive TNFR2+ TILs was ablated. Thus, TNFR2+ TILs are strongly connected to improved prognosis in TNBC; these findings suggest that TNFR2+ TILs have favorable effects in TNBC patients, unlike the tumor-promoting roles attributed to them in other cancer systems. Overall, our observations propose that the TNFR2+ TIL subset should not be targeted in the course of TNBC therapy; rather, its beneficial impacts may become into power when anti-PD-1 regimens-that may potentiate immune activities-are administered to TNBC patients.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Ductal, Breast/mortality , Lymphocytes, Tumor-Infiltrating/immunology , Receptors, Tumor Necrosis Factor, Type II/metabolism , Triple Negative Breast Neoplasms/mortality , Adult , Aged , Aged, 80 and over , Carcinoma, Ductal, Breast/immunology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Female , Follow-Up Studies , Humans , Middle Aged , Prognosis , Retrospective Studies , Survival Rate , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
3.
Appl Opt ; 57(30): 8964-8970, 2018 10 20.
Article in English | MEDLINE | ID: mdl-30461883

ABSTRACT

Cervical cancer is a type of slow-growing cancer associated with high mortality rates. Early detection can enable lifesaving early intervention. Current cervical premalignant lesion detection methods suffer from both high miss rates and excessive referrals for unnecessary biopsies. Herein, coherent light backscatter and modifications in reflected white-light spectra were measured to specifically discriminate between cervical tumors and normal squamous epithelial tissues resected from a mouse xenograft model. The combined measurements resulted in 92% sensitivity and 93% specificity in discrimination between the two tissues. These methods can be used to develop a noninvasive portable optical probe for sensitive and objective detection of precancer and cancer epithelial lesions in the cervix and other accessible epithelial tissues.


Subject(s)
Epithelial Cells/cytology , Precancerous Conditions/diagnosis , Spectrum Analysis/methods , Transplantation, Heterologous , Uterine Cervical Dysplasia/diagnosis , Uterine Cervical Neoplasms/diagnosis , Animals , Disease Models, Animal , Female , Humans , Mice , Mice, Nude , Scattering, Radiation , Sensitivity and Specificity , Tumor Cells, Cultured
4.
Front Immunol ; 8: 479, 2017.
Article in English | MEDLINE | ID: mdl-28553282

ABSTRACT

High plasticity is a hallmark of mesenchymal stem cells (MSCs), and as such, their differentiation and activities may be shaped by factors of their microenvironment. Bones, tumors, and cardiomyopathy are examples of niches and conditions that contain MSCs and are enriched with tumor necrosis factor α (TNFα) and transforming growth factor ß1 (TGFß1). These two cytokines are generally considered as having opposing roles in regulating immunity and inflammation (pro- and anti-inflammatory, respectively). Here, we performed global gene expression analysis of human bone marrow-derived MSCs and identified overlap in half of the transcriptional programs that were modified by TNFα and TGFß1. The two cytokines elevated the mRNA expression of soluble factors, including mRNAs of pro-inflammatory mediators. Accordingly, the typical pro-inflammatory factor TNFα prominently induced the protein expression levels of the pro-inflammatory mediators CCL2, CXCL8 (IL-8), and cyclooxygenase-2 (Cox-2) in MSCs, through the NF-κB/p65 pathway. In parallel, TGFß1 did not elevate CXCL8 protein levels and induced the protein expression of CCL2 at much lower levels than TNFα; yet, TGFß1 readily induced Cox-2 and acted predominantly via the Smad3 pathway. Interestingly, combined stimulation of MSCs by TNFα + TGFß1 led to a cooperative induction of all three inflammatory mediators, indicating that TGFß1 functioned as a co-inflammatory cytokine in the presence of TNFα. The cooperative activities of TNFα + TGFß1 that have led to CCL2 and CXCL8 induction were almost exclusively dependent on p65 activation and were not regulated by Smad3 or by the upstream regulator TGFß-activated kinase 1 (TAK1). In contrast, the TNFα + TGFß1-induced cooperative elevation in Cox-2 was mostly dependent on Smad3 (demonstrating cooperativity with activated NF-κB) and was partly regulated by TAK1. Studies with MSCs activated by TNFα + TGFß1 revealed that they release factors that can affect other cells in their microenvironment and induce breast tumor cell elongation, migration, and scattering out of spheroid tumor masses. Thus, our findings demonstrate a TNFα + TGFß1-driven pro-inflammatory fate in MSCs, identify specific molecular mechanisms involved, and propose that TNFα + TGFß1-stimulated MSCs influence the tumor niche. These observations suggest key roles for the microenvironment in regulating MSC functions, which in turn may affect different health-related conditions.

SELECTION OF CITATIONS
SEARCH DETAIL
...