Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Int J Cancer ; 153(3): 584-599, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-36891980

ABSTRACT

Bladder cancer is common and one of the most costly cancer forms, due to a lack of curative therapies. Recently, clinical safety and efficacy of the alpha1-oleate complex was demonstrated in a placebo-controlled study of nonmuscle invasive bladder cancer. Our study investigated if long-term therapeutic efficacy is improved by repeated treatment cycles and by combining alpha1-oleate with low-dose chemotherapy. Rapidly growing bladder tumors were treated by intravesical instillation of alpha1-oleate, Epirubicin or Mitomycin C alone or in combination. One treatment cycle arrested tumor growth, with a protective effect lasting at least 4 weeks in mice receiving 8.5 mM of alpha1-oleate alone or 1.7 mM of alpha-oleate combined with Epirubicin or Mitomycin C. Repeated treatment cycles extended protection, defined by a lack of bladder pathology and a virtual absence of bladder cancer-specific gene expression. Synergy with Epirubicin was detected at the lower alpha1-oleate concentration and in vitro, alpha1-oleate was shown to enhance the uptake and nuclear translocation of Epirubicin, by tumor cells. Effects at the chromatin level affecting cell proliferation were further suggested by reduced BrdU incorporation. In addition, alpha1-oleate triggered DNA fragmentation, defined by the TUNEL assay. The results suggest that bladder cancer development may be prevented long-term in the murine model, by alpha1-oleate alone or in combination with low-dose Epirubicin. In addition, the combination of alpha1-oleate and Epirubicin reduced the size of established tumors. Exploring these potent preventive and therapeutic effects will be of immediate interest in patients with bladder cancer.


Subject(s)
Urinary Bladder Neoplasms , Urinary Bladder , Animals , Mice , Antibiotics, Antineoplastic , Epirubicin , Mitomycin/therapeutic use , Neoplasm Recurrence, Local/pathology , Oleic Acid , Urinary Bladder/pathology , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/prevention & control , Urinary Bladder Neoplasms/pathology
2.
Crit Rev Food Sci Nutr ; 62(24): 6549-6576, 2022.
Article in English | MEDLINE | ID: mdl-33819127

ABSTRACT

BACKGROUND: Endocrine-disrupting compounds (EDCs) are ubiquitous substances that are found in our everyday lives, including pesticides, plasticizers, pharmaceutical agents, personal care products, and also in food products and food packaging. Increasing epidemiological evidence suggest that EDCs may affect the development or progression of breast cancer and consequently lead to lifelong harmful health consequences, especially when exposure occurs during early life in humans. Yet so far no appraisal of the available evidence has been conducted on this topic. OBJECTIVE: To systematically review all the available epidemiological studies about the association of the levels of environmental exposures of EDCs with breast cancer risk. METHODS: The search was performed in accordance with the PRISMA guidelines. We retrieved articles from PubMed (MEDLINE) until 10 March 2021. The key words used in this research were: "Endocrine disruptor(s)" OR "Endocrine disrupting chemical(s)" OR any of the EDCs mentioned below AND "Breast cancer" to locate all relevant articles published. We included only cohort studies and case-control studies. All relevant articles were accessed in full text and were evaluated and summarized in tables. RESULTS: We identified 131 studies that met the search criteria and were included in this systematic review. EDCs reviewed herein included pesticides (e.g. p,p'-dichlorodiphenyltrichloroethane (DDT), p,p'-dichlorodiphenyldichloroethylene (DDE), atrazine, 2,3,7,8-tetrachloridibenzo-p-dioxin (TCDD or dioxin)), synthetic chemicals (e.g. bisphenol A (BPA), phthalates, per- and polyfluoroalkyl substances (PFAS), parabens, polychlorinated biphenyls (PCBs), polybrominated diphenyl ethers (PBDEs), contraceptive pills), phytoestrogens (e.g. genistein, resveratrol), and certain mycotoxins (e.g. zearalenone). Most studies assessed environmental EDCs exposure via biomarker measurements. CONCLUSION: We identified certain EDC exposures could potentially elevate the risk of breast cancer. As majority of EDCs are highly persistent in the environment and bio-accumulative, it is essential to assess the long-term impacts of EDC exposures, especially multi-generational and transgenerational. Also, since food is often a major route of exposure to EDCs, well-designed exposure assessments of potential EDCs in food and food packing are necessary and their potential link to breast cancer development need to be carefully evaluated for subsequent EDC policy making and regulations.


Subject(s)
Endocrine Disruptors , Environmental Pollutants , Neoplasms , Pesticides , Case-Control Studies , Endocrine Disruptors/toxicity , Environmental Exposure/adverse effects , Environmental Pollutants/toxicity , Epidemiologic Studies , Humans
3.
Int J Cancer ; 147(9): 2479-2492, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32319672

ABSTRACT

Potent chemotherapeutic agents are required to counteract the aggressive behavior of cancer cells and patients often experience severe side effects, due to tissue toxicity. Our study addresses if a better balance between efficacy and toxicity can be attained using the tumoricidal complex alpha1-oleate, formed by a synthetic, alpha-helical peptide comprising the N-terminal 39 amino acids of alpha-lactalbumin and the fatty acid oleic acid. Bladder cancer was established, by intravesical instillation of MB49 cells on day 0 and the treatment group received five instillations of alpha1-oleate (1.7-17 mM) on days 3 to 11. A dose-dependent reduction in tumor size, bladder size and bladder weight was recorded in the alpha1-oleate treated group, compared to sham-treated mice. Tumor markers Ki-67, Cyclin D1 and VEGF were inhibited in a dose-dependent manner, as was the expression of cancer-related genes. Remarkably, toxicity for healthy tissue was not detected in alpha1-oleate-treated, tumor-bearing mice or healthy mice or rabbits, challenged with increasing doses of the active complex. The results define a dose-dependent therapeutic effect of alpha1-oleate in a murine bladder cancer model.


Subject(s)
Antineoplastic Agents/administration & dosage , Lactalbumin/administration & dosage , Oleic Acid/administration & dosage , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder/drug effects , Administration, Intravesical , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Cell Line, Tumor/transplantation , Disease Models, Animal , Drug Screening Assays, Antitumor , Female , Humans , Lactalbumin/chemistry , Lactalbumin/toxicity , Mice , Oleic Acid/chemistry , Oleic Acid/toxicity , Rabbits , Toxicity Tests, Subchronic , Urinary Bladder/pathology , Urinary Bladder Neoplasms/pathology
4.
Crit Rev Food Sci Nutr ; 59(20): 3320-3333, 2019.
Article in English | MEDLINE | ID: mdl-29993263

ABSTRACT

Antibiotics are a key tool used nowadays in health care industry to fight against bacterial infections; however, repeated antibiotic use or misuses, have led to bacterial resistance, causing significant threats for many people with common bacterial infections. The use of probiotics to enhance gastrointestinal health has been proposed for many years. In recent years, there has been an increasing interest in the use of probiotic bacteria as alternatives for antibiotics for preventing or treating various intestinal infections. Several important underlying mechanisms responsible for the antagonistic effects of probiotics on different microorganisms include: (1) competitive exclusion for adhesion sites and nutritional sources; (2) secretion of antimicrobial substances; (3) enhancement of intestinal barrier function; and (4) immunomodulation. However, their mode of action is not very well understood and therefore a clearer understanding of these mechanisms is necessitated. This will enable appropriate probiotic strains to be selected for particular applications and may reveal new probiotic functions. The goal of this review was to highlight some studies from literature describing the probiotic interaction with several major foodborne pathogens, as well as explore the mechanisms for such probiotic-pathogen interaction. The review will conclude by presenting future perspective and challenges of probiotic application in food products.


Subject(s)
Bacterial Infections/prevention & control , Foodborne Diseases/prevention & control , Gastrointestinal Tract/microbiology , Probiotics/therapeutic use , Anti-Bacterial Agents , Antibiosis , Humans
5.
Chem Res Toxicol ; 29(5): 823-33, 2016 05 16.
Article in English | MEDLINE | ID: mdl-27058607

ABSTRACT

Contamination of food/feedstuffs by mycotoxins is a serious problem worldwide, causing severe economic losses and serious health problems in animals/humans. Deoxynivalenol (DON) is a major mycotoxin contaminant and is known to impair intestinal barrier function. Grapes and red wine are rich in polyphenols, such as resveratrol (RES), which has striking antioxidant and anti-inflammatory activities. RES is a food-derived component; therefore, it may be simultaneously present with DON in the gastrointestinal tract. The aim of this study was to explore in vitro protective effects of RES against DON-induced intestinal damage. The results showed that RES could protect DON-induced bacteria translocation because of enhanced of intestinal barrier function by restoring the DON-induced decrease in transepithelial electrical resistance and increase in paracellular permeability. Further mechanistic studies demonstrated that RES protects against DON-induced barrier dysfunction by promoting the assembly of claudin-4 in the tight junction complex. This is probably mediated through modulation of IL-6 and IL-8 secretion via mitogen-activated protein kinase-dependent pathways. Our results imply that RES can protect against DON-induced intestinal damage and that RES may be used as a novel dietary intervention strategy to reduce DON toxicity in animals/humans.


Subject(s)
Bacterial Translocation/drug effects , Intestines/drug effects , Stilbenes/pharmacology , Trichothecenes/toxicity , Animals , Cell Line , Cytokines/metabolism , Inflammation Mediators/metabolism , Intestinal Mucosa/metabolism , Intestines/physiopathology , Resveratrol , Swine
6.
Food Chem ; 463(Pt 3): 141371, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39332376

ABSTRACT

Schisandrin B (Sch B) is a predominant bioactive lignan from the fruit of a Chinese medicine food homology plant, Schisandra chinensis. Previously, we observed potent anti-tumor effect of Sch-B in colorectal cancer (CRC) and enhanced chemotherapy efficacy with fluorouracil (5-FU). However, their bioavailability and reciprocal interactions under CRC conditions are unclear. In this study, we first compared the bioavailability, metabolism and tissue distribution of Sch-B between non-tumor-bearing and xenograft CRC tumor-bearing mice. Next, we examined SchB-5-FU interactions via investigating alterations in drug metabolism and multidrug resistance. Using a validated targeted metabolomics approach, five active metabolites, including Sch-B and fluorodeoxyuridine triphosphate, were found tumor-accumulative. Co-treatment resulted in higher levels of Sch-B and 5-FU metabolites, showing improved phytochemical and drug bioavailability. Multidrug resistance gene (MDR1) was significantly downregulated upon co-treatment. Overall, we demonstrated the potential of Sch-B to serve as a promising chemotherapy adjuvant via improving drug bioavailability and metabolism, and attenuating MDR.

7.
Cancer Med ; 13(17): e70149, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39254154

ABSTRACT

BACKGROUND: The tumoricidal complex alpha1-oleate targets bladder cancer cells, triggering rapid, apoptosis-like tumor cell death. Clinical effects of alpha1-oleate were recently observed in patients with non-muscle invasive bladder cancer (NMIBC), using a randomized, placebo-controlled study protocol. AIMS: To investigate if there are dose-dependent effects of alpha1-oleate. MATERIALS AND METHODS: Here, patients with NMIBC were treated by intravesical instillation of increasing concentrations of alpha1-oleate (1.7, 8.5, or 17 mM) and the treatment response was defined relative to a placebo group. RESULTS: Strong, dose-dependent anti-tumor effects were detected in alpha1-oleate treated patients for a combination of molecular and clinical indicators; a complete or partial response was detected in 88% of tumors treated with 8.5 mM compared to 47% of tumors treated with 1.7 mM of alpha1-oleate. Uptake of alpha1-oleate by the tumor triggered rapid shedding of tumor cells into the urine and cell death by an apoptosis-like mechanism. RNA sequencing of tissue biopsies confirmed the activation of apoptotic cell death and strong inhibition of cancer gene networks, including bladder cancer related genes. Drug-related side effects were not recorded, except for local irritation at the site of instillation. DISCUSSION AND CONCLUSIONS: These dose-dependent anti-tumor effects of alpha1-oleate are promising and support the potential of alpha1-oleate treatment in patients with NMIBC.


Subject(s)
Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/genetics , Male , Female , Aged , Middle Aged , Apoptosis/drug effects , Treatment Outcome , Dose-Response Relationship, Drug , Administration, Intravesical , Antineoplastic Agents/therapeutic use , Aged, 80 and over
8.
Sci Rep ; 14(1): 3838, 2024 02 15.
Article in English | MEDLINE | ID: mdl-38360830

ABSTRACT

Though new targeted therapies for colorectal cancer, which progresses from local intestinal tumors to metastatic disease, are being developed, tumor specificity remains an important problem, and side effects a major concern. Here, we show that the protein-fatty acid complex BAMLET (bovine alpha-lactalbumin made lethal to tumor cells) can act as a peroral treatment for colorectal cancer. ApcMin/+ mice, which carry mutations relevant to hereditary and sporadic human colorectal cancer, that received BAMLET in the drinking water showed long-term protection against tumor development and decreased expression of tumor growth-, migration-, metastasis- and angiogenesis-related genes. BAMLET treatment via drinking water inhibited the Wnt/ß-catenin and PD-1 signaling pathways and prolonged survival without evidence of toxicity. Systemic disease in the lungs, livers, spleens, and kidneys, which accompanied tumor progression, was inhibited by BAMLET treatment. The metabolic response to BAMLET included carbohydrate and lipid metabolism, which were inhibited in tumor prone ApcMin/+ mice and weakly regulated in C57BL/6 mice, suggesting potential health benefits of peroral BAMLET administration in addition to the potent antitumor effects. Together, these findings suggest that BAMLET administration in the drinking water maintains antitumor pressure by removing emergent cancer cells and reprogramming gene expression in intestinal and extra-intestinal tissues.


Subject(s)
Colorectal Neoplasms , Drinking Water , Mice , Humans , Animals , Cattle , Mice, Inbred C57BL , Signal Transduction , beta Catenin
9.
ACS Pharmacol Transl Sci ; 7(3): 863-877, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38481680

ABSTRACT

Colon cancer is among the most lethal and prevalent malignant tumors in the world, and the lack of effective therapies highlights the need for novel therapeutic approaches. Schisandrin B (Sch B), a lignan extracted from the fruit ofSchisandra chinensis, has been reported for its anticancer properties. However, to date, no studies have been done to characterize the exact molecular mechanisms underlying the antitumorigenic effects of Sch B in colon cancer. This study aimed to explore the antitumorigenic effects of Sch B in colon cancer and to understand the underlying therapeutic mechanism. A comprehensive analysis of the molecular mechanism underlying the antitumorigenic effects of Sch B on human colon cancer cells was performed using a combination of Raman spectroscopy, RNA-seq, computational docking, and molecular biological experiments. The in vivo efficacy was evaluated by a mouse xenograft model. Sch B reduced cell proliferation and triggered apoptosis in human colon cancer cell lines. Raman spectroscopy, computational, RNA-seq, and molecular and cellular studies revealed that Sch B activated unfolded protein responses by interacting with CHOP and upregulating CHOP, which thereby induced apoptosis. CHOP knockdown alleviated the Sch B-induced reduction in cell viability and apoptosis. Sch B reduced colon tumor growth in vivo. Our findings demonstrated that Sch B induced apoptosis and inhibited cell proliferation and tumor growth in vitro and in vivo. These results provided an essential background for clinical trials examining the effects of Sch B in patients with colon cancer.

10.
Appl Environ Microbiol ; 79(7): 2225-32, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23354708

ABSTRACT

Defensins are small antimicrobial peptides (AMPs) that play an important role in the innate immune system of mammals. Since the effect of mycotoxin contamination of food and feed on the secretion of intestinal AMPs is poorly understood, the aim of this study was to elucidate the individual and combined effects of four common Fusarium toxins, deoxynivalenol (DON), nivalenol (NIV), zearalenone (ZEA), and fumonisin B1 (FB1), on the mRNA expression, protein secretion, and corresponding antimicrobial effects of porcine ß-defensins 1 and 2 (pBD-1 and pBD-2) using a porcine jejunal epithelial cell line, IPEC-J2. In general, upregulation of pBD-1 and pBD-2 mRNA expression occurred following exposure to Fusarium toxins, individually and in mixtures (P < 0.05). However, no significant increase in secreted pBD-1 and pBD-2 protein levels was observed, as measured by enzyme-linked immunosorbent assay (ELISA). Supernatants from IPEC-J2 cells exposed to toxins, singly or in combination, however, possessed significantly less antimicrobial activity against Escherichia coli than untreated supernatants. When single toxins and two-toxin combinations were assessed, toxicity effects were shown to be nonadditive (including synergism, potentiation, and antagonism), suggesting interactive toxin effects when cells are exposed to mycotoxin combinations. The results show that Fusarium toxins, individually and in mixtures, activate distinct antimicrobial defense mechanisms possessing the potential to alter the intestinal microbiota through diminished antimicrobial effects. Moreover, by evaluating toxin mixtures, this improved understanding of toxin effects will enable more effective risk assessments for common mycotoxin combinations observed in contaminated food and feed.


Subject(s)
Epithelial Cells/drug effects , Fusarium/metabolism , Mycotoxins/metabolism , beta-Defensins/metabolism , Animals , Cell Line , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Swine , Transcription, Genetic
11.
Pathogens ; 13(1)2023 Dec 31.
Article in English | MEDLINE | ID: mdl-38251349

ABSTRACT

Innate immunity is essential for the anti-microbial defense, but excessive immune activation may cause severe disease. In this study, immunotherapy was shown to prevent excessive innate immune activation and restore the anti-bacterial defense. E. coli-infected Asc-/- mice develop severe acute cystitis, defined by IL-1 hyper-activation, high bacterial counts, and extensive tissue pathology. Here, the interleukin-1 receptor antagonist (IL-1RA), which inhibits IL-1 hyper-activation in acute cystitis, was identified as a more potent inhibitor of inflammation and NK1R- and substance P-dependent pain than cefotaxime. Furthermore, IL-1RA treatment inhibited the excessive innate immune activation in the kidneys of infected Irf3-/- mice and restored tissue integrity. Unexpectedly, IL-1RA also accelerated bacterial clearance from infected bladders and kidneys, including antibiotic-resistant E. coli, where cefotaxime treatment was inefficient. The results suggest that by targeting the IL-1 response, control of the innate immune response to infection may be regained, with highly favorable treatment outcomes, including infections caused by antibiotic-resistant strains.

12.
Nat Rev Urol ; 19(7): 419-437, 2022 07.
Article in English | MEDLINE | ID: mdl-35732832

ABSTRACT

Innovative solutions are needed for the treatment of bacterial infections, and a range of antibacterial molecules have been explored as alternatives to antibiotics. A different approach is to investigate the immune system of the host for new ways of making the antibacterial defence more efficient. However, the immune system has a dual role as protector and cause of disease: in addition to being protective, increasing evidence shows that innate immune responses can become excessive and cause acute symptoms and tissue pathology during infection. This role of innate immunity in disease suggests that the immune system should be targeted therapeutically, to inhibit over-reactivity. The ultimate goal is to develop therapies that selectively attenuate destructive immune response cascades, while augmenting the protective antimicrobial defence but such treatment options have remained underexplored, owing to the molecular proximity of the protective and destructive effects of the immune response. The concept of innate immunomodulation therapy has been developed successfully in urinary tract infections, based on detailed studies of innate immune activation and disease pathogenesis. Effective, disease-specific, immunomodulatory strategies have been developed by targeting specific immune response regulators including key transcription factors. In acute pyelonephritis, targeting interferon regulatory factor 7 using small interfering RNA or treatment with antimicrobial peptide cathelicidin was protective and, in acute cystitis, targeting overactive effector molecules such as IL-1ß, MMP7, COX2, cAMP and the pain-sensing receptor NK1R has been successful in vivo. Furthermore, other UTI treatment strategies, such as inhibiting bacterial adhesion and vaccination, have also shown promise.


Subject(s)
Cystitis , Pyelonephritis , Urinary Tract Infections , Anti-Bacterial Agents/therapeutic use , Cystitis/drug therapy , Humans , Immunomodulation , Pyelonephritis/drug therapy , Pyelonephritis/genetics , Pyelonephritis/microbiology , Urinary Tract Infections/drug therapy
13.
Nat Rev Urol ; 18(8): 468-486, 2021 08.
Article in English | MEDLINE | ID: mdl-34131331

ABSTRACT

The most common and lethal bacterial pathogens have co-evolved with the host. Pathogens are the aggressors, and the host immune system is responsible for the defence. However, immune responses can also become destructive, and excessive innate immune activation is a major cause of infection-associated morbidity, exemplified by symptomatic urinary tract infections (UTIs), which are caused, in part, by excessive innate immune activation. Severe kidney infections (acute pyelonephritis) are a major cause of morbidity and mortality, and painful infections of the urinary bladder (acute cystitis) can become debilitating in susceptible patients. Disease severity is controlled at specific innate immune checkpoints, and a detailed understanding of their functions is crucial for strategies to counter microbial aggression with novel treatment and prevention measures. One approach is the use of bacterial molecules that reprogramme the innate immune system, accelerating or inhibiting disease processes. A very different outcome is asymptomatic bacteriuria, defined by low host immune responsiveness to bacteria with attenuated virulence. This observation provides the rationale for immunomodulation as a new therapeutic tool to deliberately modify host susceptibility, control the host response and avoid severe disease. The power of innate immunity as an arbitrator of health and disease is also highly relevant for emerging pathogens, including the current COVID-19 pandemic.


Subject(s)
Antibodies, Viral/immunology , COVID-19/epidemiology , Immunity, Innate , SARS-CoV-2/immunology , Urinary Tract Infections/diagnosis , COVID-19/immunology , Humans , Pandemics , Severity of Illness Index , Urinary Tract Infections/epidemiology , Urinary Tract Infections/immunology
14.
Nat Biotechnol ; 39(6): 754-764, 2021 06.
Article in English | MEDLINE | ID: mdl-33574609

ABSTRACT

Is the oncogene MYC upregulated or hyperactive? In the majority of human cancers, finding agents that target c-MYC has proved difficult. Here we report specific bacterial effector molecules that inhibit cellular MYC (c-MYC) in human cells. We show that uropathogenic Escherichia coli (UPEC) degrade the c-MYC protein and attenuate MYC expression in both human cells and animal tissues. c-MYC protein was rapidly degraded by both cell-free bacterial lysates and the purified bacterial protease Lon. In mice, intravesical or peroral delivery of Lon protease delayed tumor progression and increased survival in MYC-dependent bladder and colon cancer models, respectively. These results suggest that bacteria have evolved strategies to control c-MYC tissue levels in the host and that the Lon protease shows promise for therapeutic targeting of c-MYC in cancer.


Subject(s)
Colonic Neoplasms/pathology , Genes, myc , Urinary Bladder Neoplasms/pathology , Uropathogenic Escherichia coli/enzymology , Acute Disease , Animals , Disease Models, Animal , Escherichia coli Infections/pathology , Gene Deletion , Mice , Nephritis/genetics
15.
Nat Commun ; 12(1): 3427, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34103518

ABSTRACT

Partially unfolded alpha-lactalbumin forms the oleic acid complex HAMLET, with potent tumoricidal activity. Here we define a peptide-based molecular approach for targeting and killing tumor cells, and evidence of its clinical potential (ClinicalTrials.gov NCT03560479). A 39-residue alpha-helical peptide from alpha-lactalbumin is shown to gain lethality for tumor cells by forming oleic acid complexes (alpha1-oleate). Nuclear magnetic resonance measurements and computational simulations reveal a lipid core surrounded by conformationally fluid, alpha-helical peptide motifs. In a single center, placebo controlled, double blinded Phase I/II interventional clinical trial of non-muscle invasive bladder cancer, all primary end points of safety and efficacy of alpha1-oleate treatment are reached, as evaluated in an interim analysis. Intra-vesical instillations of alpha1-oleate triggers massive shedding of tumor cells and the tumor size is reduced but no drug-related side effects are detected (primary endpoints). Shed cells contain alpha1-oleate, treated tumors show evidence of apoptosis and the expression of cancer-related genes is inhibited (secondary endpoints). The results are especially encouraging for bladder cancer, where therapeutic failures and high recurrence rates create a great, unmet medical need.


Subject(s)
Peptides/chemistry , Peptides/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Amino Acid Sequence , Apoptosis/drug effects , Cell Line, Tumor , Endocytosis/drug effects , Endpoint Determination , Gene Expression Regulation, Neoplastic/drug effects , Humans , Oleic Acids/chemistry , Peptides/pharmacology , Placebos , Protein Conformation , Proton Magnetic Resonance Spectroscopy , Thermodynamics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
16.
J Food Sci ; 83(7): 1999-2007, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29863797

ABSTRACT

This study examined modulation effects of cell-free supernatants of two commonly studied probiotic bacteria Lactobacillus rhamnosus GG (LGG) and Escherichia coli Nissle 1917 (EcN) on mucin and cytokine profiles of human intestinal epithelial HT29-MTX cells. It was found that LGG and EcN supernatants differentially modulated MUC5AC and MUC5B mRNA and protein, and total mucin-like glycoprotein secretion. Regarding modulation of cytokine profiles, LGG supernatants moderately influenced the secretion of anti-inflammatory cytokines such as interleukin (IL)-4, IL-5, and IL-10, while those of EcN exerted a broad proinflammatory effect to intestinal epithelial cells by inducing the secretion of proinflammatory cytokines such as IL-8, monocyte chemotactic protein-1, transforming growth factor α, tumor necrosis factor α, granulocyte macrophage colony-stimulating factor, and interferon γ. These results suggested that LGG and EcN might produce different bioactive products that display differential modulation of mucin and cytokines, which may contribute to intestinal health and/or defense against bacteria/pathogens. PRACTICAL APPLICATION: The results suggested that LGG and EcN might produce different bioactive products that display differential modulation of mucin and cytokines, which may contribute to intestinal health and/or defense against bacteria/pathogens.


Subject(s)
Cytokines/metabolism , Epithelial Cells/drug effects , Escherichia coli/chemistry , Intestinal Mucosa/metabolism , Intestines/drug effects , Lacticaseibacillus rhamnosus/chemistry , Mucins/metabolism , Probiotics/pharmacology , Epithelial Cells/metabolism , HT29 Cells , Humans , Interferon-gamma/metabolism , Lactobacillus/metabolism , Probiotics/chemistry , Transforming Growth Factor alpha/metabolism , Tumor Necrosis Factor-alpha/metabolism
17.
Toxicol Lett ; 281: 139-151, 2017 Nov 05.
Article in English | MEDLINE | ID: mdl-28965971

ABSTRACT

Zearalenone (ZEA) has long been recognized as a xenoestrogen, while the endocrine disrupting effects of aflatoxin B1 (AFB1) have been identified recently. Due to co-occurrence and endocrine disrupting potentials of ZEA and AFB1, it was hypothesized that co-exposure to ZEA and AFB1 might affect breast cancer cell growth. Consequently, the aim of this study was to evaluate the combined effects of ZEA and AFB1 (1nM-100nM) on cell growth and cell cycle progression, using a human breast cancer cell line MCF-7. Our results showed that ZEA and AFB1 produced significant interactive effects on cell growth, DNA synthesis and cell cycle progression. While ZEA promoted growth, DNA synthesis and cell cycle progression, AFB1 was cytotoxic and counteracted the effects of ZEA. ZEA altered the expression of several breast cancer related genes, whereas AFB1 had minimal effects on gene expression. With the use of specific inhibitors, ERα, GPER and MAPK pathways were found to be responsible for ZEA's effects on cell growth; while MAPK pathways might be involved in cytotoxic effects by AFB1. This study is first to report the effects of co-exposure of ZEA and AFB1 on breast cancer cell growth, possibly through ER dependent pathway. This suggested that endocrine-disrupting mycotoxins that co-occur in human food can interact and influence human health. Future work on interactive effects of endocrine-disrupting mycotoxins or other xenoestrogens is warranted, which will contribute to improved risk assessments.


Subject(s)
Aflatoxin B1/toxicity , Cell Cycle/drug effects , Gene Expression Regulation, Neoplastic , Zearalenone/toxicity , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Profiling , Humans , MCF-7 Cells
18.
Reprod Toxicol ; 38: 47-52, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23466516

ABSTRACT

Ochratoxin A (OTA) is a common foodborne mycotoxin. Besides its classical toxicities, it is also associated with the impairment of steroidogenesis in rats. It is hypothesized that OTA may act as an endocrine disruptor by intervening 3ß-hydroxysteroid dehydrogenase/isomerase (3ß-HSD). To address this hypothesis, human placental cells JEG-3 were used in vitro to examine the effects of short- and long-term OTA exposures on expression levels of 3ß-HSD1 and progesterone secretion at 24-96h. Results showed that both cytotoxic and non-cytotoxic levels of OTA induced 3ß-HSD1 mRNA expression by 281-378% at 72 and 96h. A significant induction (43-316%) of 3ß-HSD1 protein expression was observed at 48, 72 and 96h, and the progesterone production with the involvement of 3ß-HSD1 was significantly increased by 22-89% after 48-96h. This is the first study to demonstrate OTA up-regulates 3ß-HSD1 expression in human placental cells, indicating the potential endocrine-disrupting property of OTA.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Endocrine Disruptors/administration & dosage , Ochratoxins/administration & dosage , 17-Hydroxysteroid Dehydrogenases/genetics , Cell Line, Tumor , Female , Humans , Placenta/cytology , Pregnancy , Progesterone/metabolism , RNA, Messenger/metabolism
19.
Int J Oncol ; 36(4): 991-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20198345

ABSTRACT

Given the reported side effects associated with chemotherapy and surgical resection, dietary intervention with omega-3 polyunsaturated fatty acids (PUFAs) has been postulated to be an alterative way to prevent liver cancer progression and metastasis. We studied the effects of an omega-3 PUFA, docahexaenoic acid (DHA) on COX-2 expression and the cell cycle control machinery that co-ordinately regulate the HCC cells growth. Our data showed that DHA (0-200 microM) retarded proliferation of the human metastatic HCC cell line MHCC97L dose-dependently. In addition, inhibition of cyclin A/Cdk2 interfered with S-phase progression further in agreement with the result of bivariate flow cytometric analysis which indicated that DNA synthesis time (Ts) was significantly prolonged by DHA in MHCC97L. The N-myc oncogene, the heat shock proteins Hsp27 and glucose-related protein 78 (GRP78) as well as the antioxidant enzymes superoxide dismutase may play significant roles in the cell cycle control and reduced-proliferation of MHCC97L by DHA. Our data indicated that it is imperative to develop therapeutic strategy with omega-3 PUFA that simultaneously targets COX-2 and other cell cycle regulators in hepatocarcinogenesis. This study provides novel mechanistic insights into the modulation of DHA on human hepatocarcinoma.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/pathology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Docosahexaenoic Acids/pharmacology , Liver Neoplasms/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/secondary , Cell Adhesion/drug effects , Cell Line, Tumor , Cyclin A/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , DNA Replication/drug effects , Dose-Response Relationship, Drug , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Molecular Chaperones , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/metabolism , Superoxide Dismutase/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL