Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Publication year range
1.
Food Chem Toxicol ; 185: 114454, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38237855

ABSTRACT

Evidence suggests that meat processing and heat treatment may increase cancer risk through exposure to potentially carcinogenic compounds, polycyclic aromatic hydrocarbons (PAHs), and heterocyclic aromatic amines (HAAs). This study aims to investigate the effect of low concentrations of PAHs and HAAs (from 1 to 100 µmol/L/24h and 48h) in colorectal tumor cells (HT-29, HCT116, and LS174T) and to evaluate the effect of PAHs in the presence of inulin in mice. In vitro, the 4-PAHs have no effect on healthy colon cells but decreased the viability of the colorectal tumor cells and activated the mRNA and protein expressions of CYP1A1 and CYP1B1. In vivo, in mice with colitis induced by 3% DSS, the 4-PAHs (equimolar mix at 50,100, 150 mg/kg.bw, orally 3 times a week for 3 weeks) induced a loss of body weight and tumor formation. Inulin (10 g/L) had no effect on colon length and tumor formation. A significant decrease in the loss of b.w was observed in inulin group as compared to the fiber free group. These results underscore the importance of considering the biological association between low-dose exposure to 4-HAPs and diet-related colon tumors.


Subject(s)
Colorectal Neoplasms , Heterocyclic Compounds , Polycyclic Aromatic Hydrocarbons , Animals , Mice , Inulin/pharmacology , Amines/toxicity , Polycyclic Aromatic Hydrocarbons/toxicity , Dietary Supplements , Heterocyclic Compounds/toxicity
2.
Clin. transl. oncol. (Print) ; 25(5): 1389-1401, mayo 2023.
Article in English | IBECS | ID: ibc-219522

ABSTRACT

Background Adipose tissue is a major component of breast stroma. This study focused on delineating the effects of adipose stem cells (ASCs) derived from breast of healthy women and cancer patients with normal or tumor breast cells. Methods The ASCs were induced to differentiate into adipocytes, and the subsequent adipocyte conditioned media (ACM) were evaluated for their fatty acid profile, adipokine secretion and influence on proliferation, migration and invasion on tumoral (MCF-7 and SUM159) and normal (HMEC) human breast cell lines. Results An enrichment of arachidonic acid was observed in ACM from tumor tissues. Adipose tissues from tumor free secrete twice as much leptin than those from proximal or distal to the tumor. All ACMs display proliferative activity and favor invasiveness of SUM159 cells compared to MCF-7 and HMEC. All ACMs induced lipid droplets accumulation in MCF-7 cells and increased CD36 expression in tumor cells. Conclusion We conclude that among secreted factors analyzed, only arachidonic acid and leptin levels did discriminate ASCs from tumor-bearing and tumor-free breasts emphasizing the importance that other cell types could contribute to the adipose tissue secretome in a tumor context (AU)


Subject(s)
Humans , Female , Breast Neoplasms/pathology , Leptin/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Arachidonic Acid/pharmacology , Cell Line, Tumor , Cell Proliferation , MCF-7 Cells
3.
Clin Transl Oncol ; 25(5): 1389-1401, 2023 May.
Article in English | MEDLINE | ID: mdl-36520383

ABSTRACT

BACKGROUND: Adipose tissue is a major component of breast stroma. This study focused on delineating the effects of adipose stem cells (ASCs) derived from breast of healthy women and cancer patients with normal or tumor breast cells. METHODS: The ASCs were induced to differentiate into adipocytes, and the subsequent adipocyte conditioned media (ACM) were evaluated for their fatty acid profile, adipokine secretion and influence on proliferation, migration and invasion on tumoral (MCF-7 and SUM159) and normal (HMEC) human breast cell lines. RESULTS: An enrichment of arachidonic acid was observed in ACM from tumor tissues. Adipose tissues from tumor free secrete twice as much leptin than those from proximal or distal to the tumor. All ACMs display proliferative activity and favor invasiveness of SUM159 cells compared to MCF-7 and HMEC. All ACMs induced lipid droplets accumulation in MCF-7 cells and increased CD36 expression in tumor cells. CONCLUSION: We conclude that among secreted factors analyzed, only arachidonic acid and leptin levels did discriminate ASCs from tumor-bearing and tumor-free breasts emphasizing the importance that other cell types could contribute to the adipose tissue secretome in a tumor context.


Subject(s)
Breast Neoplasms , Leptin , Female , Humans , Leptin/metabolism , Leptin/pharmacology , Arachidonic Acid/metabolism , Arachidonic Acid/pharmacology , Breast Neoplasms/pathology , Secretome , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , MCF-7 Cells , Cell Proliferation , Culture Media, Conditioned/pharmacology , Cell Line, Tumor
4.
Cancers (Basel) ; 14(6)2022 Mar 14.
Article in English | MEDLINE | ID: mdl-35326638

ABSTRACT

Vasculogenic mimicry (VM) formed by aggressive tumor cells to create vascular networks connected with the endothelial cells, plays an important role in breast cancer progression. WISP2 has been considered as a tumor suppressor protein; however, the relationship between WISP2 and VM formation remains unclear. We used the in vitro tube formation assay and in vivo immunohistochemical analysis in a mouse model, and human breast tumors were used to evaluate the effect of WISP2 on VM formation. Here we report that WISP2 acts as a potent inhibitor of VM formation in breast cancer. Enforced expression of WISP2 decreased network formation while knockdown of WISP2 increased VM. Mechanistically, WISP2 increased retention of oncogenic activators YAP/TAZ in cytoplasm, leading to decreased expression of the angiogenic factor CYR61. Studies using an in vivo mouse model and human breast tumors confirmed the in vitro cell lines data. In conclusion, our results indicate that WISP2 may play a critical role in VM and highlight the critical role of WISP2 as a tumor suppressor.

5.
Leukemia ; 36(2): 540-548, 2022 02.
Article in English | MEDLINE | ID: mdl-34556797

ABSTRACT

Myeloma is characterized by bone lesions, which are related to both an increased osteoclast activity and a defect in the differentiation of medullary mesenchymal stem cells (MSCs) into osteoblasts. Outside the medullary environment, adipocyte-derived MSCs (ASCs) could represent a source of functional osteoblasts. However, we recently found a defect in the osteoblastic differentiation of ASCs from myeloma patients (MM-ASCs). We examined the effects of plasma from myeloma patients at diagnosis (MM-plasmas) and in complete remission (CR-plasmas) and from healthy donors on the osteoblastic differentiation of healthy donor-derived ASCs (HD-ASCs). Osteoblastogenesis in HD-ASCs was suppressed by MM-plasmas. Seven cytokines (ANG1, ENA-78, EGF, PDGF-AA/AB/BB, and TARC) were increased in MM-plasmas and separately inhibited the osteoblastic differentiation of HD-ASCs. Comparison of MM-ASCs and HD-ASCs by RNA sequencing showed that two master genes characterizing adipocyte differentiation, CD36 and PPARγ, were upregulated in MM-ASCs as compared to HD-ASCs. Finally, we demonstrated a significant increase in CD36 and PPARγ expression in HD-ASCs in the presence of MM-plasmas or the seven cytokines individually, similarly as in MM-ASCs. We conclude that specific cytokines in MM-plasmas, besides the well-known DKK1, inhibit the osteoblastic differentiation of MM- and HD-ASCs with a skewing towards adipocyte differentiation.


Subject(s)
Adipocytes/cytology , Cell Differentiation , Cytokines/pharmacology , Mesenchymal Stem Cells/cytology , Multiple Myeloma/metabolism , Osteoblasts/cytology , Stem Cells/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Case-Control Studies , Cells, Cultured , Healthy Volunteers , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Multiple Myeloma/pathology , Osteoblasts/drug effects , Osteoblasts/metabolism , Stem Cells/drug effects , Stem Cells/metabolism
6.
Cancers (Basel) ; 11(12)2019 Dec 13.
Article in English | MEDLINE | ID: mdl-31847105

ABSTRACT

Breast adiposity is correlated with body mass index, menopausal status and mammary density. We here wish to establish how these factors influence the cross-talk between breast adipocytes and normal or malignant breast cells. Adipocyte-derived stem cells (ASCs) were obtained from healthy women and classified into six distinct groups based on body mass index, menopausal status and mammary density. The ASCs were induced to differentiate, and the influence of their conditioned media (ACM) was determined. Unexpectedly, there were no detectable differences in adipogenic differentiation and secretion between the six ASC groups, while their corresponding ACMs had no detectable influence on normal breast cells. In clear contrast, all ACMs profoundly influenced the proliferation, migration and invasiveness of malignant breast cells and increased the number of lipid droplets in their cytoplasm via increased expression of the fatty acid receptor CD36, thereby increasing fatty acid uptake. Importantly, inhibition of CD36 reduced lipid droplet accumulation and attenuated the migration and invasion of the breast cancer cells. These findings suggest that breast-associated adipocytes potentiate the invasiveness of breast cancer cells which, at least in part, is mediated by metabolic reprogramming via CD36-mediated fatty acid uptake.

8.
Sci Rep ; 7(1): 1749, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28496206

ABSTRACT

WISP1 (Wnt1-inducible signaling pathway protein-1, also known as CCN4) is a member of the CCN family able to mediate cell growth, transformation and survival in a tissue-specific manner. Here, we report that WISP1 expression was highly increased in preadipocytes and decreased during adipocyte differentiation. Moreover, we observed an increase in WISP1 gene expression in adipose tissue from both diet-induced and leptin-deficient ob/ob obese mice, suggesting that WISP1 could be involved in the pathophysiological onset of obesity. Interestingly, overexpression of WISP1 in 3T3-F442A cells prevented adipocyte differentiation via downregulation of peroxisome proliferator-activated receptor (PPARγ) transcriptional activity thereby attenuating the expression of adipogenic markers. Conversely, silencing of WISP1 enhanced adipocyte differentiation. We further show that the inactivation of PPARγ transcriptional activity was mediated, at least in part, by a direct physical association between WISP1 and PPARγ, followed by proteasome-dependent degradation of PPARγ. These results suggest for the first time that WISP1 interacts with PPARγ and that this interaction results in the inhibition of PPARγ activity. Taken together our results suggest that WISP1 functions as a negative regulator of adipogenesis.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , CCN Intercellular Signaling Proteins/metabolism , Cell Differentiation , PPAR gamma/metabolism , Proto-Oncogene Proteins/metabolism , 3T3-L1 Cells , Adipogenesis/genetics , Adipose Tissue/metabolism , Animals , CCN Intercellular Signaling Proteins/genetics , Cell Differentiation/genetics , Down-Regulation/genetics , Fibroblasts/metabolism , Gene Knockdown Techniques , Gene Silencing , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Proto-Oncogene Proteins/genetics , Transcription, Genetic , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/genetics , Wnt Signaling Pathway
SELECTION OF CITATIONS
SEARCH DETAIL
...