Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38791788

ABSTRACT

Public restrooms are often a hub of microbial contamination and the examination of bacterial contamination in these facilities can serve as an important indicator of the transmission of infectious diseases. This study was conducted to determine the prevalence of bacterial contamination in public restrooms based on the economic class of the building. Samples were collected from various spots in 32 restrooms found in 10 shopping malls, classifying them into two categories: upper-end restrooms and lower-end restrooms. The findings showed that the level of contamination was higher in the lower-end restrooms, with the seat being the most contaminated area. The most dominant Gram-positive bacteria were of the coagulase-negative staphylococci species, making up 86% of the identified Gram-positive isolates. The most dominant Gram-negative bacteria identified were Klebsiella pneumoniae (K. pneumoniae) and Pseudomonas aeruginosa (P. aeruginosa). The antibiotic sensitivity test results revealed the presence of multidrug-resistant bacteria among the Gram-positive and negative isolates, including Staphylococcus haemolyticus (S. haemolyticus), Staphylococcus kloosii (S. kloosii), Acinetobacter baumanii (A. baumanii), and P. aeruginosa. In conclusion, the study underscores the significance of monitoring bacterial contamination in public restrooms and the need for measures to reduce the spread of infectious diseases. Further research is crucial to gain a complete understanding of the bacterial contamination in public restrooms and their resistance patterns, to ensure the safety and health of the public. The implementation of improved cleaning practices and hands-free designs in addition to the installation of antimicrobial surfaces in restrooms can help reduce the risk of cross-contamination and prevent the spread of diseases.


Subject(s)
Drug Resistance, Multiple, Bacterial , Bacterial Load , Toilet Facilities , Microbial Sensitivity Tests , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/isolation & purification , Humans , Bacteria/drug effects , Bacteria/isolation & purification , Bacteria/classification , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/isolation & purification , Anti-Bacterial Agents/pharmacology , Equipment Contamination
2.
J Biomol Struct Dyn ; : 1-12, 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38533896

ABSTRACT

The increasing global incidence of non-insulin-dependent diabetes mellitus (NIDDM) necessitates innovative therapeutic solutions. This study focuses on the design, synthesis and biological evaluation of Schiff base derivatives from 2-bromo-2-(2-chlorophenyl) acetic acid, particularly hydrazone compounds 4a and 4b. Both in-vitro and in-vivo assays demonstrate these derivatives' strong antidiabetic and anti-hyperlipidemic properties. In a 15-d experiment, we administered 4a and 4b at doses of 2.5 and 5 mg/kg body weight, which effectively improved symptoms of alloxan-induced diabetes in mice. These symptoms included weight loss, increased water consumption and high blood glucose levels. The compounds also normalized abnormal levels of total cholesterol (TC), triacylglycerol (TG) and low-density lipoprotein cholesterol (LDL-C), while raising the levels of high-density lipoprotein cholesterol (HDLC). Computational analysis showed that these compounds effectively inhibited the α-glucosidase enzyme by interacting with key catalytic residues, specifically Asp214 and Asp349. These computational results were confirmed through in-vitro tests, where 4a and 4b showed strong α-glucosidase inhibitory activity, with IC50 values of 0.70 ± 0.11 and 10.29 ± 0.30 µM, respectively. These compounds were more effective than the standard drug, acarbose, which had an IC50 value of 873.34 ± 1.67 µM. Mechanistic studies further indicated competitive inhibition, reinforcing the therapeutic potential of 4a and 4b for NIDDM treatment.Communicated by Ramaswamy H. Sarma.

3.
Nutrients ; 15(7)2023 Mar 29.
Article in English | MEDLINE | ID: mdl-37049499

ABSTRACT

Drug resistance is a well-known and significant obstacle in the battle against cancer, rendering chemotherapy treatments often ineffective. To improve the effectiveness of chemotherapy, researchers are exploring the use of natural molecules that can enhance its ability to kill cancer cells and limit their spread. Docosahexaenoic acid (DHA), a lipid found in marine fish, has been shown to enhance the cytotoxicity of various anti-cancer drugs in vitro and in vivo. While the combined use of chemotherapeutic drugs with DHA demonstrated promising preliminary results in clinical trials, there is still a significant amount of information to be discovered regarding the precise mechanism of action of DHA. As the biological pathways involved in the chemosensitization of already chemoresistant MCF-7 cells are still not entirely unraveled, in this study, we aimed to investigate whether DHA co-treatment could enhance the ability of the chemotherapy drug doxorubicin to inhibit the growth and invasion of MCF-7 breast cancer cells (MCF-7/Dox) that had become resistant to the drug. Upon treating MCF-7/Dox cells with DHA or DHA-doxorubicin, it was observed that the DHA-doxorubicin combination effectively enhanced cancer cell death by impeding in vitro propagation and invasive ability. In addition, it led to an increase in doxorubicin accumulation and triggered apoptosis by arresting the cell cycle at the G2/M phase. Other observed effects included a decrease in the multi-drug resistance (MDR) carrier P-glycoprotein (P-gp) and TG2, a tumor survival factor. Augmented quantities of molecules promoting apoptosis such as Bak1 and caspase-3 and enhanced lipid peroxidation were also detected. Our findings in the cell model suggest that DHA can be further investigated as a natural compound to be used alongside doxorubicin in the treatment of breast cancer that is unresponsive to chemotherapy.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Humans , Animals , Female , MCF-7 Cells , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Drug Resistance, Neoplasm , Doxorubicin/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/metabolism , Apoptosis , Cell Line, Tumor
4.
Cell Physiol Biochem ; 57(2): 105-122, 2023 Apr 12.
Article in English | MEDLINE | ID: mdl-37052042

ABSTRACT

BACKGROUND/AIMS: Macrophages interact with tumor cells within the tumor microenvironment (TME), which plays a crucial role in tumor progression. Cancer cells also can instruct macrophages to facilitate the spread of cancer and the growth of tumors. Thus, modulating macrophages-cancer cells interaction in the TME may be therapeutically beneficial. Although calcitriol (an active form of vitamin D) has anticancer properties, its role in TME is unclear. This study examined the role of calcitriol in the regulation of macrophages and cancer cells in the TME and its influence on the proliferation of breast cancer cells. METHODS: We modeled the TME, in vitro, by collecting conditioned medium from cancer cells (CCM) and macrophages (MCM) and culturing each cell type separately with and without (control) a high-dose (0.5 µM) calcitriol (an active form of vitamin D). An MTT assay was used to examine cell viability. Apoptosis was detected using FITC (fluorescein isothiocyanate) annexin V apoptosis detection kit. Western blotting was used to separate and identify proteins. Quantitative real-time PCR was used to analyze gene expression. Molecular docking studies were performed to evaluate the binding type and interactions of calcitriol to the GLUT1 and mTORC1 ligand-binding sites. RESULTS: Calcitriol treatment suppressed the expression of genes and proteins implicated in glycolysis (GLUT1, HKII, LDHA), promoted cancer cell apoptosis, and reduced viability and Cyclin D1gene expression in MCM-induced breast cancer cells. Additionally, calcitriol treatment suppressed mTOR activation in MCM-induced breast cancer cells. Molecular docking studies further showed efficient binding of calcitriol with GLUT1 and mTORC1. Calcitriol also inhibited CCM-mediated induction of CD206 and increased TNFα gene expression in THP1-derived macrophages. CONCLUSION: The results suggest that calcitriol may impact breast cancer progression by inhibiting glycolysis and M2 macrophage polarization via regulating mTOR activation in the TME and warrants further investigation in vivo.


Subject(s)
Breast Neoplasms , Calcitriol , Humans , Female , Calcitriol/pharmacology , Calcitriol/therapeutic use , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Molecular Docking Simulation , Tumor Microenvironment/genetics , TOR Serine-Threonine Kinases/metabolism , Macrophages/metabolism , Breast Neoplasms/pathology , Mechanistic Target of Rapamycin Complex 1/metabolism , Glycolysis , Cell Proliferation/genetics , Cell Line, Tumor , Macrophage Activation
5.
Antibiotics (Basel) ; 12(2)2023 Feb 08.
Article in English | MEDLINE | ID: mdl-36830265

ABSTRACT

In the current study, methanol (ADAM) extracts and their fractions, including chloroform (ADAC), ethyl acetate (ADAE), n-hexane (ADAH), and aqueous (ADAA) fractions, were prepared from aerial parts of Anogeissus dhofarica and evaluated for phytochemical assessment, high-resolution electrospray ionization mass spectrometry (HR-ESI-MS) analysis, and in vitro bioassays. The qualitative analysis determined that, except alkaloids, all the representative groups were found to be present in the analyzed samples. Samples under quantitative study displayed the highest amount of total phenolic contents in the ADAE fraction, while total flavonoid contents were highest in the ADAM extract. The ADAM extract was subjected to HR-ESI-MS to identify the chemical constituents that presented twenty-two bioactive ingredients, outlined for the first time from A. dhofarica, mainly contributed by sub-class flavanones. In the case of antimicrobial activity, the ADAE extract revealed an effective zone of inhibition (ZOI) against the Gram-positive bacterial strain (Staphylococcus aureus) with an MIC value of 0.78 ± 0.3 mg/mL, while the ADAA extract exhibited higher ZOI (34 ± 0.12 mm) against the fungal strain Candida kruzei with an MIC of 0.78 mg/mL. In the DPPH (2,2-diphenyl-1-picrylhydrazyl) analysis, the ADAE extract exhibited a maximum scavenging potential with an IC50 of 9.8 ± 1.2 µg/mL, succeeded by the ADAM extract with an IC50 of 17.4 ± 0.4 µg/mL free radical scavenging capability. In the antidiabetic assessment, the ADAE extract was the most effective, with an IC50 of 6.40 ± 0.1 µg/mL, while the same extract demonstrated prominent activity with 30.8% viability and an IC50 of 6.2 ± 0.3 µg/mL against breast cancer cell lines. The brine shrimp lethality assay demonstrated a correlation with the in vitro cytotoxicity assay, showing the ADAE extract as the most active, with a 70% mortality rate and an LC50 of 300.1 µg/mL. In conclusion, all the tested samples, especially the ADAE and ADAM extracts, have significant capabilities for the investigated activities that could be due to the presence of the bioactive compounds.

6.
Life (Basel) ; 14(1)2023 Dec 22.
Article in English | MEDLINE | ID: mdl-38255639

ABSTRACT

Cardiovascular disease (CVD) is the leading cause of death worldwide. In addition to the high mortality rate, people suffering from CVD often endure difficulties with physical activities and productivity that significantly affect their quality of life. The high prevalence of debilitating risk factors such as obesity, type 2 diabetes mellitus, smoking, hypertension, and hyperlipidemia only predicts a bleak future. Current traditional CVD interventions offer temporary respite; however, they compound the severe economic strain of health-related expenditures. Furthermore, these therapeutics can be prescribed indefinitely. Recent advances in the field of epigenetics have generated new treatment options by confronting CVD at an epigenetic level. This involves modulating gene expression by altering the organization of our genome rather than altering the DNA sequence itself. Epigenetic changes are heritable, reversible, and influenced by environmental factors such as medications. As CVD is physiologically and pathologically diverse in nature, epigenetic interventions can offer a ray of hope to replace or be combined with traditional therapeutics to provide the prospect of addressing more than just the symptoms of CVD. This review discusses various risk factors contributing to CVD, perspectives of current traditional medications in practice, and a focus on potential epigenetic therapeutics to be used as alternatives.

7.
Cancers (Basel) ; 14(20)2022 Oct 17.
Article in English | MEDLINE | ID: mdl-36291864

ABSTRACT

Imidazole-based pyrimidine hybrids are considered a remarkable class of compounds in pharmaceutical chemistry. Here, we report the anticancer bioactivities of eleven imidazole-based pyrimidine hybrids (1-11) that specifically target cytosolic carbonic anhydrase (CAs) isoenzymes, including human CA-II and human CA-IX (hCA-II, and hCA-IX). A highly eco-friendly aqueous approach was used for the formation of a carbon-carbon bond by reacting aromatic nitro group substitution of nitroimidazoles with carbon nucleophiles. The in vitro results indicate that this new class of compounds (1-11) includes significant inhibitors of hCA IX with IC50 values in the range of 9.6 ± 0.2-32.2 ± 1.0 µM, while hCA II showed IC50 values in range of 11.6 ± 0.2-31.1 ± 1.3 µM. Compound 2 (IC50 = 12.3 ± 0.1 µM) showed selective inhibition for hCA-II while 7, 8, and 10 (IC50 = 9.6-32.2 µM) were selective for hCA-IX. The mechanism of action was investigated through in vitro kinetics studies that revealed that compounds 7, 3, 11, 10, 4, and 9 for CA-IX and 1, 2, and 11 for CA-II are competitive inhibitors with dissociation constant (Ki) in the range of 7.32-17.02 µM. Furthermore, the in situ cytotoxicity of these compounds was investigated in the human breast cancer cell line MDA-MB-231 and compared with the normal human breast cell line, MCF-10A. Compound 5 showed excellent anticancer/cytotoxic activity in MDA-MB-231 with no toxicity to the normal breast cells. In addition, in silico molecular docking was employed to predict the binding mechanism of active compounds with their targets. This in silico observation aligned with our experimental results. Our findings signify that imidazole-based hybrids could be a useful choice to design anticancer agents for breast and lung tumors, or antiglaucoma compounds, by specific inhibition of carbonic anhydrases.

8.
Biomed Pharmacother ; 155: 113715, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36152415

ABSTRACT

White adipose tissue expands rapidly due to increased adipocyte number (hyperplasia) and size (hypertrophy), which results in obesity. Adipogenesis is a process of the formation of mature adipocytes from precursor cells. Additionally, obesity-related metabolic complications, such as fatty liver and insulin resistance, are linked to adipogenesis. On the contrary, autophagy is a catabolic process; essential to maintain cellular homeostasis via the degradation or recycling of unnecessary or damaged components. Importantly, autophagy dictates obesity and adipogenesis. Hence, a clear understanding of how autophagy regulates adipogenesis is crucial for drug development and the prevention and treatment of obesity and its associated disorders, such as type 2 diabetes, cardiovascular disease, and cancer. In this review, we highlighted recent findings regarding the crosstalk between adipogenesis and autophagy, as well as the molecules involved. Furthermore, the review discussed how bioactive compounds regulate adipogenesis by manipulating autophagy and underlying molecular mechanisms. Based on in vitro and animal studies, we summarized the effects of bioactive compounds on adipogenesis and autophagy. Hence, human studies are necessary to validate the effectiveness and optimal dosage of these bioactive compounds.


Subject(s)
Adipogenesis , Diabetes Mellitus, Type 2 , Animals , Humans , Adipogenesis/physiology , Diabetes Mellitus, Type 2/metabolism , Adipocytes , Autophagy , Obesity/drug therapy , Obesity/metabolism
9.
Life Sci ; 301: 120610, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35525305

ABSTRACT

AIMS: Breast cancer metastasis is the leading cause of mortality among breast cancer patients. Epithelial to mesenchymal transition (EMT) is a biological process that plays a fundamental role in facilitating breast cancer metastasis. The present study assessed the efficacy of parthenolide (PTL Tanacetum parthenium) on EMT and its underlying mechanisms in both lowly metastatic, estrogen-receptor positive, MCF-7 cells and highly metastatic, triple-negative MDA-MB-231 cells. MAIN METHODS: MCF-7 and MDA-MB-231 cells were treated with PTL (2 µM and 5 µM). Cell viability was determined by MTT (3-(4,5-dimethy lthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay. Apoptosis was analyzed by the FITC (fluorescein isothiocyanate) annexin V apoptosis detection kit. The monolayer wound scratch assay was employed to evaluate cancer cell migration. Proteins were separated and identified by Western blotting. Gene expression was analyzed by quantitative real-time PCR. KEY FINDINGS: PTL treatment significantly reduced cell viability and migration while inducing apoptosis in both cell lines. Also, PTL treatment reverses the EMT process by decreasing the mesenchymal marker vimentin and increasing the epithelial marker E-cadherin compared to the control treatment. Importantly, PTL downregulates TWIST1 (a transcription factor and regulator of EMT) gene expression, concomitant with the reduction of transforming growth factor beta1 (TGFß1) protein and gene expression in both cell lines. Additionally, molecular docking studies suggest that PTL may induce anticancer properties by targeting TGFß1 in both breast cancer cell lines. SIGNIFICANCE: Our findings provide insights into the therapeutic potential of PTL to mitigate EMT and breast cancer metastasis. These promising results demand in vivo studies.


Subject(s)
Breast Neoplasms , Epithelial-Mesenchymal Transition , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Molecular Docking Simulation , Sesquiterpenes , Transforming Growth Factor beta1/metabolism
10.
Int J Mol Sci ; 22(12)2021 Jun 18.
Article in English | MEDLINE | ID: mdl-34207035

ABSTRACT

Breast cancer is the most commonly occurring cancer in women of Western countries and is the leading cause of cancer-related mortality. The breast tumor microenvironment contains immune cells, fibroblasts, adipocytes, mesenchymal stem cells, and extracellular matrix. Among these cells, macrophages or tumor-associated macrophages (TAMs) are the major components of the breast cancer microenvironment. TAMs facilitate metastasis of the breast tumor and are responsible for poor clinical outcomes. High TAM density was also found liable for the poor prognosis of breast cancer. These observations make altering TAM function a potential therapeutic target to treat breast cancer. The present review summarizes the origin of TAMs, mechanisms of macrophage recruitment and polarization in the tumor, and the contributions of TAMs in tumor progression. We have also discussed our current knowledge about TAM-targeted therapies and the roles of miRNAs and exosomes in re-educating TAM function.


Subject(s)
Breast Neoplasms/etiology , Breast Neoplasms/metabolism , Tumor Microenvironment , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Animals , Biomarkers, Tumor , Breast Neoplasms/mortality , Breast Neoplasms/therapy , Cell Communication , Disease Progression , Disease Susceptibility , Exosomes/metabolism , Female , Gene Expression Regulation , Humans , Immunomodulation , Macrophage Activation/immunology , MicroRNAs/genetics , Neoplasm Metastasis , Neoplasm Staging , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Tumor Burden , Tumor-Associated Macrophages/pathology
11.
Sci Rep ; 11(1): 10364, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33990655

ABSTRACT

Rosiglitazone is an effective insulin-sensitizer, however associated with bone loss mainly due to increased bone resorption and bone marrow adiposity. We investigated the effect of the co-administration of fish oil rich in omega-3 fatty acids (FAs) on rosiglitazone-induced bone loss in C57BL/6 mice and the mechanisms underlying potential preventive effect. Mice fed the iso-caloric diet supplemented with fish oil exhibited significantly higher levels of bone density in different regions compared to the other groups. In the same cohort of mice, reduced activity of COX-2, enhanced activity of alkaline phosphatase, lower levels of cathepsin k, PPAR-γ, and pro-inflammatory cytokines, and a higher level of anti-inflammatory cytokines were observed. Moreover, fish oil restored rosiglitazone-induced down-regulation of osteoblast differentiation and up-regulation of adipocyte differentiation in C3H10T1/2 cells and inhibited the up-regulation of osteoclast differentiation of RANKL-treated RAW264.7 cells. We finally tested our hypothesis on human Mesenchymal Stromal Cells differentiated to osteocytes and adipocytes confirming the beneficial effect of docosahexaenoic acid (DHA) omega-3 FA during treatment with rosiglitazone, through the down-regulation of adipogenic genes, such as adipsin and FABP4 along the PPARγ/FABP4 axis, and reducing the capability of osteocytes to switch toward adipogenesis. Fish oil may prevent rosiglitazone-induced bone loss by inhibiting inflammation, osteoclastogenesis, and adipogenesis and by enhancing osteogenesis in the bone microenvironment.


Subject(s)
Bone Diseases, Metabolic/prevention & control , Dietary Supplements , Fatty Acids, Omega-3/administration & dosage , Rosiglitazone/adverse effects , Adipogenesis/drug effects , Aging/physiology , Animals , Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/physiopathology , Cell Differentiation/drug effects , Diabetes Mellitus, Type 2/drug therapy , Disease Models, Animal , Female , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoblasts/physiology , Osteoclasts/drug effects , Osteoclasts/physiology , Osteogenesis/drug effects , Primary Cell Culture , RAW 264.7 Cells
12.
Biochem J ; 478(7): 1359-1375, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33861844

ABSTRACT

Atherosclerosis is a chronic inflammatory disease associated with lipid metabolism disorder. Autophagy is a catabolic process and contributes to maintaining cellular homeostasis. Substantial evidence suggests that defective autophagy is implicated in several diseases, including atherosclerosis, while increased autophagy mitigates atherosclerosis development. Thus, understanding the mechanisms of autophagy regulation and its association with atherosclerosis is vital to develop new therapies against atherosclerosis. Dietary bioactive compounds are non-nutrient natural compounds that include phenolics, flavonoids, and carotenoids. Importantly, these bioactive compounds possess anti-inflammatory, antioxidant, and antibacterial properties that may alleviate various chronic diseases. Recently, examining the effects of bioactive compounds on autophagy activity in atherogenesis has drawn considerable attention. The current review discusses the role of macrophage autophagy in the development and progression of atherosclerosis. We also summarize our current knowledge of the therapeutic potential of bioactive compounds on atherosclerosis and autophagy.


Subject(s)
Atherosclerosis/drug therapy , Autophagy , Biological Products/pharmacology , Macrophages/pathology , Animals , Atherosclerosis/pathology , Humans
13.
Mol Biol Rep ; 47(10): 7771-7782, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32990902

ABSTRACT

Obesity is associated with hypercholesterolemia and is a global epidemic. Epidemiological and animal studies revealed cholesterol is an essential regulator of estrogen receptor positive (ER+) breast cancer progression while inhibition of cholesterol accumulation was found to prevent breast tumor growth. Individually, vitamin D and LXR agonist T0901317 showed anticancer properties. The present study investigated the effects of vitamin D3 (VD3, calcitriol), LXR agonist (T0901317) and a combination of VD3 + T0901317 on cholesterol metabolism and cancer progression in ER+ breast cancer (MCF-7) cells. VD3 or T0901317 alone reduced cholesterol accumulation significantly in MCF-7 cells concomitant with an induction of ABCA1 protein and gene expression compared to the control treatment. Most importantly, VD3 + T0901317 combination showed higher effects in reducing cholesterol levels and increasing ABCA1 protein and gene expression compared to individual treatments. Importantly, VD3 + T0901317 combination showed higher effects in increasing apoptosis as measured by annexin apoptosis assay, cell viability and was associated with induction of CHOP protein and gene expression. Additionally, the VD3 + T0901317 exerted higher effects in reducing antiapoptotic BCL-2 while increased pro-apoptotic BAX gene expression compared to the individual treatments. The present results suggest that VD3 and T0901317 combination may have an important therapeutic application to prevent obesity and hyperlipidemia mediated ER+ breast cancer progression.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Apoptosis/drug effects , Breast Neoplasms/metabolism , Cholesterol/metabolism , Hydrocarbons, Fluorinated/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , Sulfonamides/pharmacology , Transcription Factor CHOP/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Humans , Liver X Receptors/agonists , MCF-7 Cells
14.
Endocr Relat Cancer ; 26(12): R653-R672, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31525726

ABSTRACT

Obesity is a complex disease and a global epidemic. It is a risk factor for other chronic diseases including breast cancer, especially in women after menopause. Diverse etiologies underlie the relationship between obesity and breast cancer. Adipose tissue is in part responsible for these interactions. In obesity, adipose tissue undergoes several metabolic dysregulations resulting in the secretion of many pro-inflammatory cytokines, growth factors, and hormones which in turn, can promote tumor microenvironment (TME) formation and cancer progression within the breast tissue. Angiotensin II (Ang II) is a well-known hypertensive hormone produced systemically and locally by the renin-angiotensin system (RAS). Activation of this system in obesity is a potential contributor to local and systemic inflammation in breast adipose tissue. Ang II actions are primarily mediated through binding to its two receptors, type 1 (AT1R) and type 2 (AT2R). RAS inhibitors include angiotensin-converting enzyme inhibitors (ACE-I) and angiotensin receptor blockers (ARBs) which are currently prescribed as safe antihypertensive therapies. Recent studies have explored the potential use of ACE-I and ARBs in breast cancer patients as anti-tumor agents. Therefore, it is vital to understand the role of RAS in breast cancer and identify mechanisms of Ang II and RAS inhibitors in the TME and in obesity and breast cancer crosstalk. In this review, we performed a detailed analysis and discussed mechanisms of Ang II-AT1R interactions in breast cancer with emphasis on obesity-associated breast cancer. We further summarized recent in vitro, in vivo and human studies that used ACE-I/ARB interventions to improve breast cancer outcomes.


Subject(s)
Breast Neoplasms/complications , Breast Neoplasms/etiology , Obesity/complications , Obesity/etiology , Renin-Angiotensin System/genetics , Breast Neoplasms/pathology , Female , Humans , Obesity/pathology
15.
J Nutr Biochem ; 58: 1-16, 2018 08.
Article in English | MEDLINE | ID: mdl-29621669

ABSTRACT

Strategies to reduce obesity have become public health priorities as the prevalence of obesity has risen in the United States and around the world. While the anti-inflammatory and hypotriglyceridemic properties of long-chain omega-3 polyunsaturated fatty acids (n-3 PUFAs) are well known, their antiobesity effects and efficacy against metabolic syndrome, especially in humans, are still under debate. In animal models, evidence consistently suggests a role for n-3 PUFAs in reducing fat mass, particularly in the retroperitoneal and epididymal regions. In humans, however, published research suggests that though n-3 PUFAs may not aid weight loss, they may attenuate further weight gain and could be useful in the diet or as a supplement to help maintain weight loss. Proposed mechanisms by which n-3 PUFAs may work to improve body composition and counteract obesity-related metabolic changes include modulating lipid metabolism; regulating adipokines, such as adiponectin and leptin; alleviating adipose tissue inflammation; promoting adipogenesis and altering epigenetic mechanisms.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Metabolic Syndrome/diet therapy , Obesity/diet therapy , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Anti-Obesity Agents/pharmacology , Body Composition/drug effects , Exercise , Humans , Insulin Resistance , Metabolic Syndrome/prevention & control , Obesity/prevention & control , Panniculitis/diet therapy , Panniculitis/metabolism , Panniculitis/prevention & control , Vegetables/chemistry
16.
J Nutr Biochem ; 53: 111-120, 2018 03.
Article in English | MEDLINE | ID: mdl-29216499

ABSTRACT

Breast cancer is one of the major causes of death in the USA. Cancer cells, including breast, have high glycolysis rates to meet their energy demands for survival and growth. Vitamin D3 (VD3) is important for many important physiological processes such as bone mineralization, but its anticancer role is yet to be proven. We find that VD3 treatment significantly down-regulates glycolytic enzymes and genes and decreases glucose uptake - for both lowly metastatic MCF-7 and highly metastatic MDA-MB-231 (MB231) breast cancer cells. VD3 also significantly decreases cell viability by inducing apoptosis - consistent with decreased expression of mammalian target of rapamycin (mTOR), which regulates glycolysis and cancer cell survival, and increases 5' adenosine monophosphate-activated protein kinase (AMPK) activation. These changes accompany a significant reduction of cell migration and increased cell stiffness, presumably a consequence of reversal of the epithelial to mesenchymal transition resulting in increased E-cadherin, and F-actin, and reduced vimentin expression. High levels of cytoskeletal and cortical F-actin may cause high cell stiffness. VD3-induced mechanical changes are stronger in highly metastatic MB231 than in lowly metastatic MCF-7 cells. Our results suggest therapeutic and preventive roles of VD3 in breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cholecalciferol/pharmacology , Glycolysis/drug effects , Apoptosis/drug effects , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Enzymes/genetics , Enzymes/metabolism , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Glucose/pharmacokinetics , Glycolysis/physiology , Humans , Lactic Acid/metabolism , MCF-7 Cells , TOR Serine-Threonine Kinases/metabolism
17.
JCI Insight ; 1(18): e88646, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27812542

ABSTRACT

Emerging evidence indicates that upregulation of the ER stress-induced pro-osteogenic transcription factor ATF4 plays an important role in vascular calcification, a common complication in patients with aging, diabetes, and chronic kidney disease (CKD). In this study, we demonstrated the pathophysiological role of ATF4 in vascular calcification using global Atf4 KO, smooth muscle cell-specific (SMC-specific) Atf4 KO, and transgenic (TG) mouse models. Reduced expression of ATF4 in global ATF4-haplodeficient and SMC-specific Atf4 KO mice reduced medial and atherosclerotic calcification under normal kidney and CKD conditions. In contrast, increased expression of ATF4 in SMC-specific Atf4 TG mice caused severe medial and atherosclerotic calcification. We further demonstrated that ATF4 transcriptionally upregulates the expression of type III sodium-dependent phosphate cotransporters (PiT1 and PiT2) by interacting with C/EBPß. These results demonstrate that the ER stress effector ATF4 plays a critical role in the pathogenesis of vascular calcification through increased phosphate uptake in vascular SMCs.


Subject(s)
Activating Transcription Factor 4/genetics , Myocytes, Smooth Muscle/metabolism , Vascular Calcification/metabolism , Animals , Cells, Cultured , Humans , Ion Pumps/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Muscle, Smooth , Muscle, Smooth, Vascular/cytology , Vascular Calcification/pathology
18.
J Oleo Sci ; 65(7): 613-20, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27321121

ABSTRACT

Chlorella (Parachlorella beijerinckii) powder is reported to show a preventive effect against metabolic syndromes such as arteriosclerosis, hyperlipidemia, and hypertension. Approximately 60% of the chlorella content is protein. In order to understand the role of chlorella protein, we prepared a chlorella protein hydrolysate (CPH) by protease treatment. Male C57BL/6 mice were divided into three groups: a normal diet group, high-fat diet (HFD) group, and high-fat diet supplemented with CPH (HFD+CPH) group. The CPH administration improved glucose intolerance, insulin sensitivity, and adipose tissue hypertrophy in the high-fat diet-fed mice. In addition, the HFD+CPH group had significantly decreased liver total cholesterol and triglyceride levels compared with those in the HFD group. Furthermore, the HFD+CPH group had a decreased level of monocyte chemotactic protein-1 (MCP-1) in serum and a lower MCP-1 mRNA expression level in adipose tissue compared with the HFD group. The present study suggests that chlorella protein hydrolysate can prevent a high-fat diet-induced glucose disorder and fatty liver by inhibiting adipocyte hypertrophy and reducing the MCP-1 protein and gene expression.


Subject(s)
Chlorella/chemistry , Diet, High-Fat/adverse effects , Fatty Liver/drug therapy , Glucose Metabolism Disorders/drug therapy , Obesity/metabolism , Protein Hydrolysates/chemistry , Protein Hydrolysates/pharmacology , Animals , Chlorella/metabolism , Fatty Liver/metabolism , Glucose Metabolism Disorders/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/etiology
19.
J Biol Chem ; 287(44): 37340-51, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22955269

ABSTRACT

Chronic hepatitis C virus (HCV) infection greatly increases the risk for type 2 diabetes and nonalcoholic steatohepatitis; however, the pathogenic mechanisms remain incompletely understood. Here we report gluconeogenic enzyme phosphoenolpyruvate carboxykinase (PEPCK) transcription and associated transcription factors are dramatically up-regulated in Huh.8 cells, which stably express an HCV subgenome replicon. HCV increased activation of cAMP response element-binding protein (CREB), CCAAT/enhancer-binding protein (C/EBPß), forkhead box protein O1 (FOXO1), and peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) and involved activation of the cAMP response element in the PEPCK promoter. Infection with dominant-negative CREB or C/EBPß-shRNA significantly reduced or normalized PEPCK expression, with no change in PGC-1α or FOXO1 levels. Notably, expression of HCV nonstructural component NS5A in Huh7 or primary hepatocytes stimulated PEPCK gene expression and glucose output in HepG2 cells, whereas a deletion in NS5A reduced PEPCK expression and lowered cellular lipids but was without effect on insulin resistance, as demonstrated by the inability of insulin to stimulate mobilization of a pool of insulin-responsive vesicles to the plasma membrane. HCV-replicating cells demonstrated increases in cellular lipids with insulin resistance at the level of the insulin receptor, increased insulin receptor substrate 1 (Ser-312), and decreased Akt (Ser-473) activation in response to insulin. C/EBPß-RNAi normalized lipogenic genes sterol regulatory element-binding protein-1c, peroxisome proliferator-activated receptor γ, and liver X receptor α but was unable to reduce accumulation of triglycerides in Huh.8 cells or reverse the increase in ApoB expression, suggesting a role for increased lipid retention in steatotic hepatocytes. Collectively, these data reveal an important role of NS5A, C/EBPß, and pCREB in promoting HCV-induced gluconeogenic gene expression and suggest that increased C/EBPß and NS5A may be essential components leading to increased gluconeogenesis associated with HCV infection.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/physiology , Fatty Liver/virology , Genome, Viral , Hepacivirus/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Viral Nonstructural Proteins/physiology , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/metabolism , Diabetes Mellitus, Type 2/virology , Enzyme Induction , Fatty Liver/enzymology , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Genes, Reporter , Gluconeogenesis/genetics , Glucose/metabolism , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Hepacivirus/physiology , Humans , Insulin/physiology , Lipid Metabolism/genetics , Luciferases/biosynthesis , Luciferases/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Promoter Regions, Genetic , Rats , Secretory Vesicles/metabolism , Signal Transduction , Virus Replication
20.
Mol Cell Endocrinol ; 331(1): 102-8, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20797423

ABSTRACT

Diabetes and obesity are associated with activation of endoplasmic reticulum (ER) stress; however a direct link between ER stress and increased hepatic gluconeogenesis remains unclear. Here we show that ER stress triggers a significant increase in expression of CCAAT/enhancer-binding protein (C/EBPß) and phosphorylated CREB together with reduced phospho-AMP-activated protein kinase (pAMPK) in hepatoma cells. ER stress contributed to transcriptional activation of the gluconeogenic phosphoenolpyruvate carboxykinase (PEPCK) promoter in Huh7 and HepG2 cells via cAMP binding motif (CRE site). Chromatin immunoprecipitation assays demonstrate that C/EBPß is recruited to the PEPCK promoter during ER stress and is reversed by pre-treatment with a JNK inhibitor that relieves ER stress. C/EBPß but not pCREB was suppressed by the AMPK-activator AICAR or constitutively active AMPK, while dominant negative AMPK increased C/EBPß expression. These data suggest that ER stress triggers suppression of AMPK while increasing C/EBPß and pCREB expression which activates PEPCK gene transcription. Understanding how ER stress suppresses AMPK activation and increases C/EBPß expression could lead to a potentially novel pathway for treatment of diabetes.


Subject(s)
Adenylate Kinase/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Carcinoma, Hepatocellular/genetics , Endoplasmic Reticulum/pathology , Liver Neoplasms/genetics , Phosphoenolpyruvate Carboxykinase (ATP)/genetics , Up-Regulation/genetics , Adenylate Kinase/antagonists & inhibitors , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/metabolism , Endoplasmic Reticulum/genetics , Enzyme Activation/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Models, Genetic , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Protein Binding/drug effects , Protein Kinase Inhibitors/pharmacology , Rats , Response Elements/genetics , Stress, Physiological/drug effects , Stress, Physiological/genetics , Thapsigargin/pharmacology , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...