Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Biomed Rep ; 19(2): 56, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37560313

ABSTRACT

Breast cancer (BC) is the most common cancer in women worldwide, with 2.3 million cases recorded in 2020. Despite improvements in cancer treatment, patients with BC still succumb to the disease, due to regional and distant metastases when diagnosed at later stages. Several immune checkpoint inhibitors have been approved for BC treatment, based on their expression and role in maintaining immunosurveillance against tumors. The present study aimed to evaluate the expression of 12 immune checkpoints in patients with BC, and assess their role as diagnostic and therapeutic markers. Expression levels were measured using reverse transcription-quantitative polymerase chain reaction. Among the 12 immune markers, herpesvirus entry mediator (HVEM) was found to be significantly upregulated in patients with malignant BC compared to non-malignant controls, with a relative fold change (FC) of 1.46 and P=0.012. A similar finding was observed for cytotoxic T-lymphocyte-associated antigen 4 (CTLA4; FC=1.47 and P=0.035). In addition, receiver operating characteristic curve analysis revealed that HVEM expression allowed significant differentiation between groups, with an area under the curve of 0.74 (P=0.013). Upregulation in both HVEM and CTLA4 was revealed to be significantly associated with the human epidermal growth factor receptor-2 (HER2)-enriched phenotype (FC=3.53, P=0.009 and FC=5.98, P=0.002, respectively), while only HVEM was significantly associated with the triple-negative phenotype (FC=2.07, P=0.016). Furthermore, HVEM was significantly higher in patients with grade III tumors (FC=1.88, P=0.025) and negative vascular invasion (FC=1.67, P=0.046) compared with non-malignant controls. Serum protein levels were assessed by multiplex immunoassay, and a significant increase in HVEM was detected in patients with malignant BC compared with that in non-malignant controls (P=0.035). These data indicated that HVEM may serve as a potential biomarker and target for immunotherapy, especially for certain types of BC.

2.
Molecules ; 27(9)2022 Apr 27.
Article in English | MEDLINE | ID: mdl-35566130

ABSTRACT

Nigella sativa oil, commonly known as black seed oil (BSO), is a well-known Mediterranean food, and its consumption is associated with beneficial effects on human health. A large number of BSO's therapeutic properties is attributed to its pharmacologically active compound, thymoquinone (TQ), which inhibits cell proliferation and induces apoptosis by targeting several epigenetic players, including the ubiquitin-like, containing plant homeodomain (PHD) and an interesting new gene, RING finger domains 1 (UHRF1), and its partners, DNA methyltransferase 1 (DNMT1) and histone deacetylase 1 (HDAC1). This study was designed to compare the effects of locally sourced BSO with those of pure TQ on the expression of the epigenetic complex UHRF1/DNMT1/HDAC1 and the related events in several cancer cells. The gas chromatographs obtained from GC-MS analyses of extracted BSO showed that TQ was the major volatile compound. BSO significantly inhibited the proliferation of MCF-7, HeLa and Jurkat cells in a dose-dependent manner, and it induced apoptosis in these cell lines. BSO-induced inhibitory effects were associated with a significant decrease in mRNA expression of UHRF1, DNMT1 and HDAC1. Molecular docking and MD simulation showed that TQ had good binding affinity to UHRF1 and HDAC1. Of note, TQ formed a stable metal coordinate bond with zinc tom, found in the active site of the HDAC1 protein. These findings suggest that the use of TQ-rich BSO represents a promising strategy for epigenetic therapy for both solid and blood tumors through direct targeting of the trimeric epigenetic complex UHRF1/DNMT1/ HDAC1.


Subject(s)
Neoplasms , Nigella sativa , Benzoquinones/pharmacology , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Epigenesis, Genetic , Humans , Molecular Docking Simulation , Neoplasms/drug therapy , Neoplasms/genetics , Nigella sativa/metabolism , Plant Oils/pharmacology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
3.
Oncol Lett ; 22(1): 508, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33986869

ABSTRACT

Long non-coding RNAs (lncRNAs), a type of cellular RNA, play a critical regulatory role in several physiological developments and pathological processes, such as tumorigenesis and tumor progression. Obesity is a risk factor for a number of serious health conditions, including breast cancer (BC). However, the underlying mechanisms behind the association between obesity and increased BC incidence and mortality remain unclear. Several studies have reported changes in lncRNA expression due to obesity and BC, independently encouraging further investigation of the relationship between the two in connection with lncRNAs. The present study was designed to first screen for the expression of 29 selected lncRNAs that showed a link to cancer or obesity in the blood of a selected cohort of 6 obese and 6 non-obese patients with BC. The expression levels of significantly expressed lncRNAs, AP001429.1, PCAT6, P5549, P19461 and P3134, were further investigated in a larger cohort of 69 patients with BC (36 obese and 33 non-obese), using reverse transcription-quantitative polymerase chain reaction. Results showed not only that AP001429.1 remained significantly downregulated in the larger cohort (P=0.002), but also that it was associated with several clinicopathological characteristics, such as negative HER2 status, negative E-cadherin expression, negative vascular invasion, negative margin invasion and LCIS. These findings suggest that obesity may have a role in inhibiting AP001429.1 expression, which may serve as a novel potential biomarker and therapeutic target for BC.

4.
Front Oncol ; 10: 804, 2020.
Article in English | MEDLINE | ID: mdl-32509585

ABSTRACT

Information regarding transcriptome and metabolome has significantly contributed to identifying potential therapeutic targets for the management of a variety of cancers. Obesity has profound effects on both cancer cell transcriptome and metabolome that can affect the outcome of cancer therapy. The information regarding the potential effects of obesity on breast cancer (BC) transcriptome, metabolome, and its integration to identify novel pathways related to disease progression are still elusive. We assessed the whole blood transcriptome and serum metabolome, as circulating metabolites, of obese BC patients compared them with non-obese BC patients. In these patients' samples, 186 significant differentially expressed genes (DEGs) were identified, comprising 156 upregulated and 30 downregulated. The expressions of these gene were confirmed by qRT-PCR. Furthermore, 96 deregulated metabolites were identified as untargeted metabolomics in the same group of patients. These detected DEGs and deregulated metabolites enriched in many cellular pathways. Further investigation, by integration analysis between transcriptomics and metabolomics data at the pathway levels, revealed seven unique enriched pathways in obese BC patients when compared with non-obese BC patients, which may provide resistance for BC cells to dodge the circulating immune cells in the blood. In conclusion, this study provides information on the unique pathways altered at transcriptome and metabolome levels in obese BC patients that could provide an important tool for researchers and contribute further to knowledge on the molecular interaction between obesity and BC. Further studies are needed to confirm this and to elucidate the exact underlying mechanism for the effects of obesity on the BC initiation or/and progression.

5.
BMC Womens Health ; 17(1): 105, 2017 Nov 09.
Article in English | MEDLINE | ID: mdl-29121911

ABSTRACT

BACKGROUND: Obesity is part of the established risk factors for breast cancer (BC) in postmenopausal females. Circulating leptin increases in parallel with the increase of body weight and fat reservoir. METHODS: This research investigated the link between leptin phenotype and the clinicopathological factors in BC. A large set of breast cancer cases (449), and 27 non-cancerous tissue samples of breast were employed for leptin expression recognition using immunohistochemistry staining. RESULTS: Cytoplasmic immunohistochemical staining of leptin was recognized in 376 (83.7%) and 25 (92.6%) of BC and control cases respectively. Leptin immunostaining were significantly associated with age, histotypes, grade, stage, lymph node involvement, tumor recurrence, hormone receptor phenotypes, ER and HER2 expressions, and p-values were (P = 0.0233), (P = 0.0001), (P = 0.050), (P = 0.0291), (P = 0.0300), (P = 0.0023), (P = 0.0021), (P = 0.0279) respectively. Reasonable proportion of cases with low staining score was more prevalent in all subgroups of clinicopathological parameters except ER- PR+ HER2- hormone receptor phenotype and mucinous carcinoma which showed high level of leptin immunoreactivity. Tumor recurrence is less prevailing in high score leptin immunostaining cases. Furthermore, Log Rank (Mantel-Cox) test findings revealed considerably different survival distributions were observed for the different categories of leptin immunostaining scores (P = 0.032). Negative leptin immunostaining is related to poor survival. CONCLUSIONS: Our preliminary findings support leptin clinical value in confirming BC diagnosis as well as prognosis. These results suggest that leptin molecule is an important biomarker that could identify type, grade, stage, lymph node involvement, relapse and prognosis in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Leptin/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Female , Humans , Immunohistochemistry , Lymph Nodes/pathology , Prognosis , Staining and Labeling
6.
Ann Diagn Pathol ; 29: 62-67, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28807345

ABSTRACT

Many investigators have examined the functions of AMP-activated protein kinase (AMPK) in cancer biology and its anti-neoplastic features in cancer models. The goal of this research is to assess the association of the immunohistochemical expression of AMPK in human mammary tumours with the clinical data of breast cancer patients. 449 cases of previously diagnosed breast cancer, and 27 tissue samples of fibroadenomas and normal breast were utilized for detection of AMPK expression using tissue microarrays and immunohistochemistry. Brownish nuclear and cytoplasmic staining were present in epithelial cells and stromal cells in 333 (74.16%) and 348 (77.5%) cancer cases respectively indicating AMPK expression. Twenty two (81.48%) control cases showed AMPK immunoexpression in both epithelial and stromal cells. Significant statistical association has been found between advanced stages of breast cancer and increased intensity of AMPK immunostaining only in epithelial cells (p-value=0.0001). Histotypes have been correlated with AMPK immunostaining in epithelial cells only (p-value=0.029). Low AMPK immunostaining scores were more dominant in DCIS, ductal and mixed type's ductal and mucinous histotypes, while high intense staining was more common in the lobular type. Furthermore, breast tumour cases with lymph node metastases showed significant AMPK expression in both epithelial and stromal cells (p-value=0.0001 and p-value=0.026). Low scores of AMPK immunostaining were common in breast cancer cases with positive vascular invasion (p-value=0.007) and disease recurrence (p-value=0.008). No significant differences in survival behavior distributions were observed for the different categories of AMPK immunostaining in epithelial and stromal cells. In conclusion, our results showed decreased AMPK expression in breast cancer in comparison with the control group. AMPK expression was significantly correlated with some clinicopathological factors like advanced stage, lymph node involvement, vascular invasion and disease recurrence which give indications for poor clinical outcomes. Immunohistochemical staining of AMPK protein is a valuable method which could predict cases of breast cancer with poor prognosis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Neoplasm Recurrence, Local/pathology , Adult , Aged , Aged, 80 and over , Carcinoma, Ductal, Breast/diagnosis , Female , Fibroadenoma/pathology , Humans , Immunohistochemistry/methods , Lymphatic Metastasis , Middle Aged , Neoplasm Recurrence, Local/diagnosis , Prognosis , Stromal Cells/metabolism , Stromal Cells/pathology , Young Adult
7.
Oncol Rep ; 34(4): 1675-80, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26260992

ABSTRACT

AMP-activated protein kinase (AMPK), a sensor of cellular energy, is widely reported as a potential therapeutic target in treatment of breast and other cancers. The activated enzyme has been shown to be a promising anti-proliferative agent in breast cancer cell lines. However, little data exist on crosstalk between AMPK and the cellular survival axis of PI3K/Akt/mTOR pathway and the impact of microenvironment on cellular responses to AMPK activation. We present results which show differential crosstalk between AMPK and Akt, dependent on the cellular genetics of each breast cancer cell type. We also show that leptin blocks activation of AMPK and partially or completely attenuates the anti-proliferative effect of AMPK activation depending on the cell type. This suggests that leptin within the local environment might impose limitations on therapeutic usage of AMPK activators in cancer, thereby attenuating their effective use in many obese subjects.


Subject(s)
AMP-Activated Protein Kinases/genetics , Breast Neoplasms/genetics , Enzyme Activation/genetics , Leptin/genetics , AMP-Activated Protein Kinases/biosynthesis , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Genotype , Humans , Leptin/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , TOR Serine-Threonine Kinases/genetics , Tumor Microenvironment/genetics
8.
Int J Cancer ; 123(7): 1536-44, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18634052

ABSTRACT

New efforts are being focused on signalling pathways as targets for cancer therapy. This particular study was designed to investigate whether blockade of the phosphatidylinositol 3OH-kinase (PI3K) pathway (a survival/anti-apoptosis pathway, overexpressed in various tumours) could sensitise human breast cancer cells to the effect of chemotherapeutics. Doxorubicin (Dox) and LY294002 (LY, a PI3K inhibitor) were used individually or in combination on MDA-MB-231 (p53 mutant, ER-), T47D (p53 mutant, ER+), and MCF-7 (p53 wildtype, ER+) human breast cancer cell lines, and on 184A1, a nonmalignant human breast epithelial cell line (p53 wildtype, ER-). Each drug showed time- and dose-dependent growth inhibition of cell proliferation on all 4 cell lines. The combination of Dox+LY resulted in enhanced cell growth inhibition in MDA-MB-231 and T47D cells, and additive inhibition in MCF-7 and 184A1 cells. Cell cycle analysis showed that Dox+LY enhanced the arrest of MDA-MB-231 and T47D cells in G2 with the appearance of a sub-G1 peak indicating apoptosis/necrosis, a notion supported by enhanced depolarisation of mitochondrial membrane potential in these cell types. The combination also caused a greater additive increase in Cyclin B1. Thus, the synergistic effect of the combination on cell proliferation in some, but not all, breast cancer cells may be through enhanced induction of both G2 arrest and apoptosis, in which p53 may play a role. Substantially lower doses of doxorubicin could be used with low doses of inhibitors of the PI3K pathway, without compromising the anti-cancer effect, but also lowering detrimental side-effects of doxorubicin. This study supports the notion that survival signalling pathways offer special targets for chemotherapy in cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Doxorubicin/pharmacology , Enzyme Inhibitors/pharmacology , Mammary Glands, Human/drug effects , Phosphoinositide-3 Kinase Inhibitors , Receptors, Estrogen/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Cycle , Cell Line , Cell Line, Tumor , Cell Proliferation , Chromones/pharmacology , Flow Cytometry , Humans , Mammary Glands, Human/metabolism , Membrane Potentials , Mitochondria/drug effects , Mitochondria/physiology , Morpholines/pharmacology
9.
Eur Urol ; 43(3): 301-8, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12600435

ABSTRACT

OBJECTIVES: Recent studies suggest a paracrine/autocrine loop involving prolactin (PRL) within the human prostate. The aims of this study were to determine the effects of PRL on the growth and survival of prostate cancer cells and the intracellular signalling mechanisms underlying such effects. METHODS: The effect of PRL on proliferation of LNCaP, PC3 and DU145 was assessed by Coulter counting. The effect of PRL on TRAIL-, staurosporine- and flavopiridol-induced apoptosis was assessed by Timelapse microscopy and Annexin V binding. The status of the PRL receptor (PRL-R) and Akt/PKB (protein kinase B) activity were assessed by Western blotting. RESULTS: All three cell lines expressed both the short and long forms of the PRL receptor. Although, no significant effect of PRL on the proliferation of these cells was found, PRL partially inhibited TRAIL-induced apoptosis in PC3 cells. PRL also enhanced the phosphorylation of Akt/PKB in these cells. CONCLUSIONS: PRL had no significant effect on the proliferation of PC3, DU145 and LNCaP, but inhibited TRAIL-induced apoptosis in PC3 cells, possibly via enhanced Akt/PKB phosphorylation in PC3 cells. Further investigations are underway to determine the survival effect of PRL on the other two prostate cancer cell line.


Subject(s)
Apoptosis/drug effects , Prolactin/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases , Signal Transduction/drug effects , Apoptosis/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Survival/drug effects , Cell Survival/physiology , Humans , Male , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins c-akt , Receptors, Prolactin/drug effects , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...