Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Methods ; 229: 30-40, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38880434

ABSTRACT

The objective of this study was to optimise the millet formulation using Levilactobacillus brevis and to evaluate its anticarcinogenic potential in vitro. The formula was developed in the course of the fermentation of finger millet (Eleusine coracana) using L. brevis MTTC 4460 and optimised by response surface methodology and validation by artificial neural networking (ANN). The optimised millet formulation could be obtained using 2 % of bacterial inoculum, 2 % of glucose, and a fermentation duration of 3.3 days with a yield of 5.98 mg/mL lactic acid and 3.38 log10 (CFU/mL) viable L. brevis with overall desirability value of 1. The fermented millet formulation exhibited antiproliferative and antimigratory effects on MDA-MB-231 and HCT116 cancer cell lines. In addition, the outcomes observed in western blot analysis revealed that the formulation elicited apoptotic responses mediated by the Bcl-2 family of proteins in MDA-MB-231 and HCT116 cell lines while demonstrating no discernible impact on HEK293 normal cells.

2.
FEBS Lett ; 598(7): 818-836, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38418371

ABSTRACT

Plasmodium falciparum renovates the host erythrocyte to survive during intraerythrocytic development. This renovation requires many parasite proteins to unfold and move outside the parasitophorous vacuolar membrane, and chaperone-regulated protein folding becomes essential for the exported proteins to function. We report on a type-IV J domain protein (JDP), PF3D7_1401100, which we found to be processed before export and trafficked inside the lumen of parasite-derived structures known as J-dots. We found this protein to have holdase activity, as well as stimulate the ATPase and aggregation suppression activity of the human HSP70 chaperone HsHSPA8; thus, we named it "HSPA8-interacting J protein" (A8iJp). Moreover, we found a subset of HsHSPA8 to co-localize with A8iJp inside the infected human erythrocyte. Our results suggest that A8iJp modulates HsHSPA8 chaperone activity and may play an important role in host erythrocyte renovation.


Subject(s)
HSP40 Heat-Shock Proteins , Plasmodium falciparum , Humans , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/chemistry , HSP40 Heat-Shock Proteins/metabolism , Protein Binding , Protozoan Proteins/metabolism , Molecular Chaperones/metabolism , Erythrocytes , Protein Folding , HSC70 Heat-Shock Proteins/metabolism
3.
Med Oncol ; 41(3): 78, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38393411

ABSTRACT

Breast cancer progression, metastasis and recurrence are largely driven by breast cancer stem cells (BCSCs), which constitute a subset of tumor cells exhibiting stem cell characteristics. In this study, we evaluated the role of estrogen-related receptor alpha (ERRα) in the migration, invasion and angiogenesis of BCSCs. The inhibition of ERRα using XCT790 or knockdown of ERRα using shRNA inhibited the mammosphere formation efficiency, as well as the migration and invasion of BCSCs derived from the mammospheres of MCF7 and MDA-MB-231 (MB231) cells. Conversely, the overexpression of ERRα significantly increased the migration and invasion of BCSCs derived from the mammosphere. In addition, the XCT790 treatment or shERRα significantly downregulated the epithelial-mesenchymal transition (EMT), as evidenced by the downregulation in the expression of vimentin, Snail, Slug and N-cadherin in the mammospheres of MCF7 and MB231 cells. The chorioallantoic membrane assay showed that the conditioned media from XCT790-treated and shERRα cells significantly inhibited blood vessel formation and vessel length. Furthermore, XCT790 treatment or shERRα also downregulated the expression of molecular markers of angiogenesis, such as VEGF-A and Ang-2 in the mammospheres. Conversely, the overexpression of ERRα in MCF7 cells significantly increased both EMT and angiogenesis. These findings suggest that ERRα inhibits the migration, invasion and angiogenesis of BCSCs, suggesting as a potential target for breast cancer therapy.


Subject(s)
Breast Neoplasms , ERRalpha Estrogen-Related Receptor , Nitriles , Thiazoles , Female , Humans , Angiogenesis , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition/genetics , Neoplastic Stem Cells/pathology , Receptors, Estrogen/metabolism
4.
Med Oncol ; 40(11): 326, 2023 Oct 08.
Article in English | MEDLINE | ID: mdl-37806999

ABSTRACT

Breast cancer is the most prevalent form of cancer in women globally, and TNBC (triple-negative breast cancer) is its aggressive type since it lacks the usual targets. JAK2/STAT3 pathway can be an important lead in anticancer drug discovery, as restraining the downstream signalling of this pathway results in the induction of cell apoptosis. Moreover, various limitations associated with chemotherapy are the reason to find an alternative herbal-based therapy. For this study, we collected Urtica dioica and U. parviflora from different regions of Uttarakhand, followed by preparation of their leaf and stem extracts in different solvents. The GC-MS analysis of these extracts revealed a total of 175 compounds to be present in them. Further, by molecular docking approach, we studied the interaction between these compounds and JAK2, and 12 major compounds with better binding energy than the control Paclitaxel were identified. In addition, the selected hits were also reported to display better pharmacokinetic properties. Moreover, extracts from both the Urtica spp. displayed significant anticancer activity against MDA-MB-231(TNBC cell line) and exhibited lower cytotoxicity in healthy cell lines, i.e. HEK293T, indicating that these extracts were safer to use. Hence, the findings in our study can be crucial in the area of herbal-based target-specific drug development against breast cancer.


Subject(s)
Triple Negative Breast Neoplasms , Female , Humans , Triple Negative Breast Neoplasms/metabolism , Molecular Docking Simulation , HEK293 Cells , Cell Line, Tumor , Paclitaxel/therapeutic use , Apoptosis , Cell Proliferation , Janus Kinase 2
5.
Stem Cell Rev Rep ; 19(8): 2807-2819, 2023 11.
Article in English | MEDLINE | ID: mdl-37584854

ABSTRACT

Cancer stem cells drive tumor initiation, invasion, metastasis and recurrence. In the present study, we have evaluated the role of ERRα in the maintenance of breast cancer stem cells (BCSCs) using breast cancer cell lines. The inhibition of ERRα with the inverse agonist, XCT-790, or the knockdown of ERRα in breast cancer cells significantly reduced the mammosphere formation efficiency and mammosphere size along with a significant reduction in the CD44+/CD24- BCSCs. Treatment with XCT-790 significantly downregulated expression of the transcription factors involved in stem cell maintenance such as Oct4, Klf4, Sox2, Nanog and c-Myc in the mammosphere forming stem cells of MCF7 and MDA-MB-231. In addition, XCT-790 induced cell cycle arrest and apoptosis in the mammosphere-forming cells. The knockdown or inhibition of ERRα downregulated the expression of Notch1 and ß-catenin, whereas the overexpression of ERRα in MCF7 cells upregulated the expression of these proteins. Moreover, the inhibition of ERRα synergistically enhanced the efficacy of paclitaxel in inhibiting the BCSCs. These results show that ERRα is crucial for the maintenance of BCSCs and suggest that ERRα could be a potential target for breast cancer treatment.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Drug Inverse Agonism , Neoplastic Stem Cells/metabolism , ERRalpha Estrogen-Related Receptor
6.
Genes (Basel) ; 14(5)2023 05 02.
Article in English | MEDLINE | ID: mdl-37239397

ABSTRACT

BACKGROUND: The high prevalence and rapid emergence of antibiotic resistance in high-risk Klebsiella pneumoniae (KP) ST147 clones is a global health concern and warrants molecular surveillance. METHODS: A pangenome analysis was performed using publicly available ST147 complete genomes. The characteristics and evolutionary relationships among ST147 members were investigated through a Bayesian phylogenetic analysis. RESULTS: The large number of accessory genes in the pangenome indicates genome plasticity and openness. Seventy-two antibiotic resistance genes were found to be linked with antibiotic inactivation, efflux, and target alteration. The exclusive detection of the blaOXA-232 gene within the ColKp3 plasmid of KP_SDL79 suggests its acquisition through horizontal gene transfer. The association of seventy-six virulence genes with the acrAB efflux pump, T6SS system and type I secretion system describes its pathogenicity. The presence of Tn6170, a putative Tn7-like transposon in KP_SDL79 with an insertion at the flanking region of the tnsB gene, establishes its transmission ability. The Bayesian phylogenetic analysis estimates ST147's initial divergence in 1951 and the most recent common ancestor for the entire KP population in 1621. CONCLUSIONS: Present study highlights the genetic diversity and evolutionary dynamics of high-risk clones of K. pneumoniae. Further inter-clonal diversity studies will help us understand its outbreak more precisely and pave the way for therapeutic interventions.


Subject(s)
Klebsiella Infections , beta-Lactamases , Humans , beta-Lactamases/genetics , Klebsiella pneumoniae/genetics , Phylogeny , Bayes Theorem , Klebsiella Infections/epidemiology , Klebsiella Infections/genetics , Klebsiella Infections/drug therapy
7.
IEEE J Biomed Health Inform ; 27(6): 2782-2793, 2023 06.
Article in English | MEDLINE | ID: mdl-37023159

ABSTRACT

During COVID-19 pandemic qRT-PCR, CT scans and biochemical parameters were studied to understand the patients' physiological changes and disease progression. There is a lack of clear understanding of the correlation of lung inflammation with biochemical parameters available. Among the 1136 patients studied, C-reactive-protein (CRP) is the most critical parameter for classifying symptomatic and asymptomatic groups. Elevated CRP is corroborated with increased D-dimer, Gamma-glutamyl-transferase (GGT), and urea levels in COVID-19 patients. To overcome the limitations of manual chest CT scoring system, we segmented the lungs and detected ground-glass-opacity (GGO) in specific lobes from 2D CT images by 2D U-Net-based deep learning (DL) approach. Our method shows accuracy, compared to the manual method (  âˆ¼ 80%), which is subjected to the radiologist's experience. We determined a positive correlation of GGO in the right upper-middle (0.34) and lower (0.26) lobe with D-dimer. However, a modest correlation was observed with CRP, ferritin and other studied parameters. The final Dice Coefficient (or the F1 score) and Intersection-Over-Union for testing accuracy are 95.44% and 91.95%, respectively. This study can help reduce the burden and manual bias besides increasing the accuracy of GGO scoring. Further study on geographically diverse large populations may help to understand the association of the biochemical parameters and pattern of GGO in lung lobes with different SARS-CoV-2 Variants of Concern's disease pathogenesis in these populations.


Subject(s)
COVID-19 , Deep Learning , Humans , COVID-19/diagnostic imaging , SARS-CoV-2 , Pandemics , Retrospective Studies , Lung/diagnostic imaging
8.
FEBS Lett ; 596(1): 95-111, 2022 01.
Article in English | MEDLINE | ID: mdl-34890056

ABSTRACT

Renovation of host erythrocytes is vital for pathogenesis by Plasmodium falciparum. These changes are mediated by parasite proteins that translocate beyond the parasitophorous vacuolar membrane in an unfolded state, suggesting protein folding by chaperones is imperative for the functionality of exported proteins. We report a type IV P. falciparum heat-shock protein 40, PF11_0034, that localizes to the cytoplasmic side of J-dots and interacts with the erythrocyte cytoskeleton, and therefore named eCiJp (erythrocyte cytoskeleton-interacting J protein). Recombinant eCiJp binds to the human heat-shock protein 70 HsHSPA1 and promotes its ATPase activity. In addition, eCiJp could suppress protein aggregation. Our data suggest that eCiJp recruits HsHSPA1 to the host erythrocyte cytoskeleton, where it may become involved in remodeling of the erythrocyte cytoskeleton and/or folding of exported parasite proteins.


Subject(s)
HSP40 Heat-Shock Proteins
9.
Mol Cell Biochem ; 476(12): 4363-4385, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34453645

ABSTRACT

Cervical cancer (CC) is one of the leading causes of death in women due to cancer and a major concern in the developing world. Persistent human papilloma virus (HPV) infection is the major causative agent for CC. Besides HPV infection, genetic and epigenetic factors including microRNA (miRNA) also contribute to the malignant transformation. Earlier studies have revealed that miRNAs participate in cell proliferation, invasion and metastasis, angiogenesis, and chemoresistance processes by binding and inversely regulating the target oncogenes or tumor suppressor genes. Based on functions and mechanistic insights, miRNAs have been identified as cellular modulators that have an enormous role in diagnosis, prognosis, and cancer therapy. Signatures of miRNA could be used as diagnostic markers which are necessary for early diagnosis and management of CC. The therapeutic potential of miRNAs has been shown in CC; however, more comprehensive clinical trials are required for the clinical translation of miRNA-based diagnostics and therapeutics. Understanding the molecular mechanism of miRNAs and their target genes has been useful to develop miRNA-based therapeutic strategies for CC and overcome chemoresistance. In this review, we summarize the role of miRNAs in the development, progression, and metastasis of CC as well as chemoresistance. Further, we discuss the diagnostic and therapeutic potential of miRNAs to overcome chemoresistance and treatment of CC.


Subject(s)
MicroRNAs/genetics , Papillomaviridae/genetics , Papillomavirus Infections/genetics , Uterine Cervical Neoplasms/pathology , Drug Resistance, Neoplasm , Female , Humans , Papillomaviridae/isolation & purification , Papillomaviridae/pathogenicity , Papillomavirus Infections/metabolism , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Prognosis , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/virology
10.
Eur J Pharmacol ; 871: 172938, 2020 Mar 15.
Article in English | MEDLINE | ID: mdl-31958458

ABSTRACT

Radiotherapy is routinely used in the treatment of breast cancer. However, its efficiency is often limited by the development of radioresistance and metastasis. The cancer cells surviving irradiation show epithelial-mesenchymal transition (EMT) along with increased migration, invasion and metastasis. In this study, we have evaluated the role of α-lipoic acid in preventing the radiation-induced EMT and in sensitizing the breast cancer cells to radiation. The breast cancer cell lines, MCF-7 and MDA-MB-231 were pretreated with lipoic acid, irradiated and the changes associated with cell growth, clonogenicity, migration, matrix metalloproteinases (MMPs), EMT and TGFß signaling were measured. Our data showed that lipoic acid pretreatment sensitized the breast cancer cells to the ionizing radiation and inhibited the radiation-induced migration and the release of MMP2 and MMP9. Lipoic acid also prevented the TGFß1 release and inhibited the radiation-induced EMT in breast cancer cells. The inhibition of TGFß signaling by lipoic acid is associated with the inhibition of radiation-induced activation and translocation of NF-κB. These results suggest that α-lipoic acid inhibits the radiation-induced TGFß signaling and nuclear translocation of NF-κB, thereby inhibiting the radiation-induced EMT and sensitizing the breast cancer cells to ionizing radiation.


Subject(s)
Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/radiation effects , Radiation Tolerance/drug effects , Thioctic Acid/pharmacology , Cell Movement/drug effects , Cell Movement/radiation effects , Humans , MCF-7 Cells , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , NF-kappa B/metabolism , Radiation Tolerance/radiation effects , Signal Transduction/drug effects , Signal Transduction/radiation effects , Transforming Growth Factor beta/metabolism
11.
Life Sci ; 207: 15-22, 2018 Aug 15.
Article in English | MEDLINE | ID: mdl-29802942

ABSTRACT

AIMS: Invasion and metastasis are the main cause of mortality in breast cancer. Hence, novel therapeutic interventions with high specificity toward invasion and metastasis are necessary. α-Lipoic acid showed antiproliferative and cytotoxic effects on several cancers including breast cancer. However, the effect of lipoic acid on breast cancer metastasis remains unclear. MAIN METHODS: In the present study, we examined the effects of lipoic acid on the migration and invasion of MDA-MB-231 and 4T1 breast cancer cells. KEY FINDINGS: Our data showed that lipoic acid effectively inhibited the colony forming ability of highly invasive MDA-MB-231 and 4T1 cells. Moreover, the nontoxic concentrations of lipoic acid significantly reduced the migration of breast cancer cells. Lipoic acid also inhibited the TGFß-induced angiopoietin-like 4 (ANGPTL4) expression and reduced the activity of matrix metalloproteinase-9 (MMP-9), an enzyme involved in invasion and metastasis, in both the cell lines. The inhibition of cell migration by lipoic acid is accompanied by the downregulation of FAK, ERK1/2 and AKT phosphorylation, and inhibition of nuclear translocation of ß-catenin. SIGNIFICANCE: Our data demonstrated that lipoic acid inhibited the migration and invasion of metastatic breast cancer cells at least in part through inhibiting ERK1/2 and AKT signaling. Thus, our findings show that the inhibition of TGFß signaling is a potential mechanism for the anti-invasive effects of lipoic acid.


Subject(s)
Breast Neoplasms/pathology , Thioctic Acid/pharmacology , Transforming Growth Factor beta1/metabolism , Active Transport, Cell Nucleus , Angiopoietin-Like Protein 4/metabolism , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement , Cell Proliferation , Dose-Response Relationship, Drug , Female , Humans , Matrix Metalloproteinase 9/metabolism , Mice , Neoplasm Invasiveness , Phosphorylation , Signal Transduction
12.
Nat Commun ; 6: 6910, 2015 Apr 24.
Article in English | MEDLINE | ID: mdl-25908435

ABSTRACT

Mammary stem/progenitor cells (MaSCs) maintain self-renewal of the mammary epithelium during puberty and pregnancy. DNA methylation provides a potential epigenetic mechanism for maintaining cellular memory during self-renewal. Although DNA methyltransferases (DNMTs) are dispensable for embryonic stem cell maintenance, their role in maintaining MaSCs and cancer stem cells (CSCs) in constantly replenishing mammary epithelium is unclear. Here we show that DNMT1 is indispensable for MaSC maintenance. Furthermore, we find that DNMT1 expression is elevated in mammary tumours, and mammary gland-specific DNMT1 deletion protects mice from mammary tumorigenesis by limiting the CSC pool. Through genome-scale methylation studies, we identify ISL1 as a direct DNMT1 target, hypermethylated and downregulated in mammary tumours and CSCs. DNMT inhibition or ISL1 expression in breast cancer cells limits CSC population. Altogether, our studies uncover an essential role for DNMT1 in MaSC and CSC maintenance and identify DNMT1-ISL1 axis as a potential therapeutic target for breast cancer treatment.


Subject(s)
Breast Neoplasms/genetics , Carcinogenesis/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , LIM-Homeodomain Proteins/genetics , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/genetics , Neoplastic Stem Cells/metabolism , Transcription Factors/genetics , Animals , Blotting, Western , Breast Neoplasms/metabolism , Cell Line , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Down-Regulation , Female , Humans , LIM-Homeodomain Proteins/metabolism , MCF-7 Cells , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Mammary Neoplasms, Experimental/metabolism , Mice , Microscopy, Fluorescence , Neoplastic Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/metabolism
13.
Cancer Res ; 74(4): 1166-78, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24371223

ABSTRACT

GPR109A, a G-protein-coupled receptor, is activated by niacin and butyrate. Upon activation in colonocytes, GPR109A potentiates anti-inflammatory pathways, induces apoptosis, and protects against inflammation-induced colon cancer. In contrast, GPR109A activation in keratinocytes induces flushing by activation of Cox-2-dependent inflammatory signaling, and the receptor expression is upregulated in human epidermoid carcinoma. Thus, depending on the cellular context and tissue, GPR109A functions either as a tumor suppressor or a tumor promoter. However, the expression status and the functional implications of this receptor in the mammary epithelium are not known. Here, we show that GPR109A is expressed in normal mammary tissue and, irrespective of the hormone receptor status, its expression is silenced in human primary breast tumor tissues, breast cancer cell lines, and in tumor tissues of three different murine mammary tumor models. Functional expression of this receptor in human breast cancer cell lines decreases cyclic AMP production, induces apoptosis, and blocks colony formation and mammary tumor growth. Transcriptome analysis revealed that GPR109A activation inhibits genes, which are involved in cell survival and antiapoptotic signaling, in human breast cancer cells. In addition, deletion of Gpr109a in mice increased tumor incidence and triggered early onset of mammary tumorigenesis with increased lung metastasis in MMTV-Neu mouse model of spontaneous breast cancer. These findings suggest that GPR109A is a tumor suppressor in mammary gland and that pharmacologic induction of this gene in tumor tissues followed by its activation with agonists could be an effective therapeutic strategy to treat breast cancer.


Subject(s)
Breast Neoplasms/genetics , Genes, Tumor Suppressor/physiology , Receptors, G-Protein-Coupled/physiology , Receptors, Nicotinic/physiology , Animals , Butyrates/metabolism , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Female , HEK293 Cells , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Niacin/metabolism
14.
Mol Cell Biol ; 33(19): 3920-35, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23918800

ABSTRACT

SLC5A8 is a putative tumor suppressor that is inactivated in more than 10 different types of cancer, but neither the oncogenic signaling responsible for SLC5A8 inactivation nor the functional relevance of SLC5A8 loss to tumor growth has been elucidated. Here, we identify oncogenic HRAS (HRAS(G12V)) as a potent mediator of SLC5A8 silencing in human nontransformed normal mammary epithelial cell lines and in mouse mammary tumors through DNMT1. Further, we demonstrate that loss of Slc5a8 increases cancer-initiating stem cell formation and promotes mammary tumorigenesis and lung metastasis in an HRAS-driven murine model of mammary tumors. Mammary-gland-specific overexpression of Slc5a8 (mouse mammary tumor virus-Slc5a8 transgenic mice), as well as induction of endogenous Slc5a8 in mice with inhibitors of DNA methylation, protects against HRAS-driven mammary tumors. Collectively, our results provide the tumor-suppressive role of SLC5A8 and identify the oncogenic HRAS as a mediator of tumor-associated silencing of this tumor suppressor in mammary glands. These findings suggest that pharmacological approaches to reactivate SLC5A8 expression in tumor cells have potential as a novel therapeutic strategy for breast cancer treatment.


Subject(s)
Breast Neoplasms/genetics , Cation Transport Proteins/genetics , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cation Transport Proteins/metabolism , Cell Line , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Female , HCT116 Cells , Humans , Immunoblotting , MCF-7 Cells , Male , Mice , Mice, Knockout , Mice, Nude , Mice, Transgenic , Monocarboxylic Acid Transporters , Mutation , Proto-Oncogene Proteins p21(ras)/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
15.
Biochem J ; 450(1): 169-78, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23167260

ABSTRACT

SLC5A8 (solute carrier gene family 5A, member 8) is a sodium-coupled transporter for monocarboxylates. Among its substrates are the HDAC (histone deacetylase) inhibitors butyrate, propionate and pyruvate. Expression of SLC5A8 is silenced in cancers via DNA methylation, and ectopic expression of SLC5A8 in cancer cells induces apoptosis in the presence of its substrates that are HDAC inhibitors. In the present study we show that ectopic expression of SLC5A8 in cancer cells translocates the anti-apoptotic protein survivin to the plasma membrane through protein-protein interaction resulting in depletion of nuclear survivin and also decreases cellular levels of survivin through inhibition of transcription. These SLC5A8-induced changes in the location and levels of survivin result in cell-cycle arrest, disruption of the chromosome passenger complex involved in mitosis, induction of apoptosis and enhancement in chemosensitivity. These effects are seen independently of the transport function of SLC5A8 and histone acetylation status of the cell; in the presence of pyruvate, a SLC5A8 substrate and also an HDAC inhibitor, these effects are amplified. Ectopic expression of SLC5A8 in the breast cancer cell line MB231 inhibits the ability of cells to form colonies in vitro and to form tumours in mouse xenografts in vivo. The suppression of survivin transcription occurs independently of HDAC inhibition, and the underlying mechanism is associated with decreased phosphorylation of STAT3 (signal transducer and activator of transcription 3). The observed effects are specific for survivin with no apparent changes in expression of other inhibitor-of-apoptosis proteins. The present study unravels a novel, hitherto unrecognized, mechanism for the tumour-suppressive role of a plasma membrane transporter independent of its transport function.


Subject(s)
Breast Neoplasms/pathology , Cation Transport Proteins/metabolism , Cell Membrane/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Pancreatic Neoplasms/pathology , Animals , Apoptosis , Biological Transport , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cation Transport Proteins/genetics , Cell Line, Tumor , Female , Humans , Inhibitor of Apoptosis Proteins/genetics , Mice , Mice, Nude , Monocarboxylic Acid Transporters , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Survivin , Transplantation, Heterologous
16.
Invest Ophthalmol Vis Sci ; 53(4): 2208-17, 2012 Apr 24.
Article in English | MEDLINE | ID: mdl-22427566

ABSTRACT

PURPOSE: Retinal pigment epithelium (RPE) expresses GPR109A, a receptor for the vitamin niacin and the ketone body ß-hydroxybutyrate (ß-HB). Because diabetes results in elevated levels of ß-HB, here we studied expression of the receptor in diabetic retina. We also investigated its functional relevance in RPE. METHODS: Retinal expression of GPR109A in diabetic mice and postmortem human eyes was evaluated by quantitative PCR (qPCR). ARPE-19 cells and primary wild-type and Gpr109a(-/-) mouse RPE cells were exposed to TNF-α in the presence or absence of niacin or ß-HB, followed by analysis of IL-6 and Ccl2 expression via real-time qPCR and ELISA. RESULTS: GPR109A expression was increased in diabetic mouse and human retina. TNF-α increased the expression and secretion of IL-6 and Ccl2 in ARPE-19 cells. Niacin and ß-HB suppressed these effects, implicating GPR109A as the target responsible for mediation of the observed effects. Primary RPE cells from wild-type mice behaved similarly. In contrast, GPR109A ligands failed to suppress TNF-α-induced expression and secretion of IL-6 and Ccl2 in primary RPE cells from Gpr109a(-/-) mice, confirming that the observed anti-inflammatory effects were mediated specifically by Gpr109a. CONCLUSIONS: GPR109A plays an anti-inflammatory role in RPE and its expression is upregulated in diabetes. Inflammation is a key causative factor in the pathogenesis of diabetic retinopathy. We speculate that the increased expression of GPR109A and elevation of its ligand ß-HB in diabetes are mechanisms by which the tissue attempts to fight inflammation in this disease. Pharmacological activation of GPR109A may therefore have therapeutic potential in clinical management of diabetic retinopathy.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Diabetic Retinopathy/genetics , Gene Expression Regulation/physiology , Receptors, G-Protein-Coupled/genetics , Receptors, Nicotinic/genetics , Retinal Pigment Epithelium/metabolism , 3-Hydroxybutyric Acid/pharmacology , Aged , Animals , Cell Line , Chemokine CCL2/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetic Retinopathy/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Niacin/pharmacology , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Retina/drug effects , Retina/metabolism , Retinal Pigment Epithelium/drug effects , Tumor Necrosis Factor-alpha/pharmacology
17.
Cancer Res ; 71(21): 6654-64, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21920899

ABSTRACT

The NAD-dependent histone deacetylase silent information regulator 1 (SIRT1) is overexpressed and catalytically activated in a number of human cancers, but recent studies have actually suggested that it may function as a tumor suppressor and metastasis inhibitor in vivo. In breast cancer, SIRT1 stabilization has been suggested to contribute to the oncogenic potential of the estrogen receptor α (ERα), but SIRT1 activity has also been associated with ERα deacetylation and inactivation. In this study, we show that SIRT1 is critical for estrogen to promote breast cancer. ERα physically interacted and functionally cooperated with SIRT1 in breast cancer cells. ERα also bound to the promoter for SIRT1 and increased its transcription. SIRT1 expression induced by ERα was sufficient to activate antioxidant and prosurvival genes in breast cancer cells, such as catalase and glutathione peroxidase, and to inactivate tumor suppressor genes such as cyclin G2 (CCNG2) and p53. Moreover, SIRT1 inactivation eliminated estrogen/ERα-induced cell growth and tumor development, triggering apoptosis. Taken together, these results indicated that SIRT1 is required for estrogen-induced breast cancer growth. Our findings imply that the combination of SIRT1 inhibitors and antiestrogen compounds may offer more effective treatment strategies for breast cancer.


Subject(s)
Breast Neoplasms/physiopathology , Estrogen Receptor alpha/physiology , Estrogens/physiology , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/physiopathology , Signal Transduction/physiology , Sirtuin 1/physiology , Acetylation , Animals , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Apoptosis/physiology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cells, Cultured , Epithelial Cells/metabolism , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor Modulators/therapeutic use , Estrogen Receptor alpha/chemistry , Female , Gene Expression Regulation, Neoplastic , Glutathione Peroxidase/biosynthesis , Glutathione Peroxidase/genetics , Humans , Lipid Peroxidation , Mice , Mice, Nude , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Protein Interaction Mapping , Protein Processing, Post-Translational , Sirtuin 1/chemistry , Specific Pathogen-Free Organisms , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
18.
J Biol Chem ; 286(36): 31830-8, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21771784

ABSTRACT

SLC6A14, also known as ATB(0,+), is an amino acid transporter with unique characteristics. It transports 18 of the 20 proteinogenic amino acids. However, this transporter is expressed only at low levels in normal tissues. Here, we show that the transporter is up-regulated specifically in estrogen receptor (ER)-positive breast cancer, demonstrable with primary human breast cancer tissues and human breast cancer cell lines. SLC6A14 is an estrogen/ER target. The transport features of SLC6A14 include concentrative transport of leucine (an activator of mTOR), glutamine (an essential amino acid for nucleotide biosynthesis and substrate for glutaminolysis), and arginine (an essential amino acid for tumor cells), suggesting that ER-positive breast cancer cells up-regulate SLC6A14 to meet their increased demand for these amino acids. Consequently, treatment of ER-positive breast cancer cells in vitro with α-methyl-DL-tryptophan (α-MT), a selective blocker of SLC6A14, induces amino acid deprivation, inhibits mTOR, and activates autophagy. Prolongation of the treatment with α-MT causes apoptosis. Addition of an autophagy inhibitor (3-methyladenine) during α-MT treatment also induces apoptosis. These effects of α-MT are specific to ER-positive breast cancer cells, which express the transporter. The ability of α-MT to cause amino acid deprivation is significantly attenuated in MCF-7 cells, an ER-positive breast cancer cell line, when SLC6A14 is silenced with shRNA. In mouse xenograft studies, α-MT by itself is able to reduce the growth of the ER-positive ZR-75-1 breast cancer cells. These studies identify SLC6A14 as a novel and effective drug target for the treatment of ER-positive breast cancer.


Subject(s)
Amino Acid Transport Systems, Neutral/antagonists & inhibitors , Breast Neoplasms/drug therapy , Amino Acid Transport Systems , Amino Acid Transport Systems, Neutral/genetics , Animals , Autophagy/drug effects , Breast Neoplasms/pathology , Female , Humans , Mice , Molecular Targeted Therapy/methods , Receptors, Estrogen , Transplantation, Heterologous , Tryptophan/analogs & derivatives , Tryptophan/pharmacology , Tumor Cells, Cultured
19.
Anticancer Res ; 30(10): 4169-76, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21036737

ABSTRACT

Tocotrienols belong to the vitamin E family of chemicals known to have potent anti-proliferative and apoptotic activities against a variety of cancer cells with little to no comparable influence on the normal cells. Whether tocotrienols control the expression of phase II antioxidant enzymes in the context of their anti-carcinogenic mechanisms has not been investigated. The present studies were performed to test whether the differential growth inhibition resulting from exposure to α-, γ- and δ-tocotrienols in estrogen receptor-positive human MCF-7 and estrogen receptor-negative MDA-MB-231 breast cancer cells might be accompanied by changes in phase II antioxidant enzymes. Cell proliferation and clonogenicity in both cell lines were significantly inhibited by γ- and δ-tocotrienols with little affect when cells were similarly exposed to α-tocotrienol, at doses up to 10 µM. The expression and activity of several antioxidant enzymes in 10 µM tocotrienol-treated cells were determined by Western blot and biochemical assays. In MDA-MB-231 cells, δ- was more active than α- or γ-tocotrienols in up-regulating glutathione peroxidase; however, the three tocotrienols had comparable activity in inducing thioredoxin. In MCF-7 cells, expression of quinone reductase 2 and thioredoxin was increased by γ- and δ-tocotrienols, whereas quinone reductase 1 was unaffected by exposure to the tocotrienols. The tocotrienols also did not affect the expression and activity of superoxide dismutase in both MCF-7 and MDA-MB-231 cells, but increased catalase activity concomitant with slight reduction in the catalase expression. In MDA-MB-231 cells, treatment by tocotrienols led to several fold increase of NRF2 expression marked by corresponding decrease in KEAP1 levels. By contrast, no significant change in NRF2 and KEAP1 levels was observed in MCF-7 cells. These studies demonstrate that different tocotrienols show distinct and selective activity in regulating the NRF2-KEAP1, in coordination with the induced expression of cytoprotective oxidative stress modulatory genes and regulation of proliferation in breast cancer cells.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Oxidative Stress/drug effects , Tocotrienols/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Catalase/biosynthesis , Cell Growth Processes/drug effects , Cell Line, Tumor , Enzyme Induction/drug effects , Female , Glutathione Peroxidase/biosynthesis , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , NF-E2-Related Factor 2/metabolism , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/metabolism , Superoxide Dismutase/biosynthesis , Superoxide Dismutase-1
20.
Anticancer Res ; 30(7): 2869-74, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20683025

ABSTRACT

BACKGROUND: Tocotrienols, a subgroup of the vitamin E family, have demonstrated antioxidant and anticancer properties. Differential growth responses among different types of tocotrienols have been observed in breast cancer cells; however, specific bioactivity of each individual tocotrienol remains to be elucidated. MATERIALS AND METHODS: In this study, the effects of gamma-tocotrienol were examined with regard to its ability to suppress cell proliferation via modulation of cell cycle regulatory protein expression, and also from the perspective of control of cellular oxidoreductive status through regulation of detoxification enzymes, e.g., quinone reductase NQO2, using estrogen receptor-positive MCF-7 human breast cancer cells. RESULTS: It was shown that treatment by gamma-tocotrienol suppressed MCF-7 cell proliferation in a dose- and time-dependent manner. Growth suppression by gamma-tocotrienol was accompanied by changes in the levels of cell cycle regulatory proteins, notably, Rb/E2F complex, cyclin D1/cdk4 and cyclin B1/cdk1, as exemplified by loss of cyclin D1, inhibition of specific Rb phosphorylation (pRb-p at Thr821), and by the time- and dose-dependent increase in the expression of NQO2. CONCLUSION: By exerting control on expression of specific cell cycle regulatory proteins in concomitance with suppression of cell proliferation, as well as the induction of NQO2, gamma-tocotrienol offers promise as an added chemopreventive and/or chemotherapeutic agent against breast cancer carcinogenesis.


Subject(s)
Breast Neoplasms/metabolism , Cell Cycle Proteins/biosynthesis , Chromans/pharmacology , Quinone Reductases/biosynthesis , Vitamin E/analogs & derivatives , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Growth Processes/drug effects , Cell Line, Tumor , Enzyme Induction/drug effects , Gene Expression/drug effects , Humans , Quinone Reductases/genetics , Up-Regulation/drug effects , Vitamin E/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...