Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 369
1.
Mol Nutr Food Res ; 68(9): e2300703, 2024 May.
Article En | MEDLINE | ID: mdl-38676329

Botanicals and herbal supplements contain a diverse array of polyphenols that may affect mammary gland function and promote galactagogue activity. This scoping review is conducted to identify scientific literature elucidating how polyphenols affect mammary gland biology and cellular mechanisms critical for lactation. A literature search of PubMed and Medline reviews relevant studies in dairy animals, rodent models, and cultured mammary epithelial cells that are published from January 2010 until July 2023, to ascertain effects of polyphenols on mechanisms regulating milk production and composition. The PRISMA-ScR (Preferred Reporting Items for Systematic Reviews and Meta-Analyses for Scoping Review) strategy is applied and 80 studies on polyphenols and their implications on milk production and composition are included in this review. Limited information delineating effects of polyphenols on the molecular pathways that affect lactation are found, although available information suggests modulation of Stat5 signaling/differentiation, Stat3 signaling/remodeling, mTOR and insulin signaling/energy production, and nuclear factor kappa beta (NFκß) signaling/oxidative stress and inflammation may play roles. A profound lack of mechanistic information underscores the critical need for further research to understand the impact of botanical supplements and polyphenols on milk production and composition in humans to establish maternal nutritional guidelines to support lactation and breastfeeding goals.


Galactogogues , Lactation , Polyphenols , Lactation/drug effects , Polyphenols/pharmacology , Female , Humans , Galactogogues/pharmacology , Animals , Dietary Supplements , Mammary Glands, Animal/drug effects , Signal Transduction/drug effects , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism
2.
Proc Natl Acad Sci U S A ; 119(11): e2115308119, 2022 03 15.
Article En | MEDLINE | ID: mdl-35263230

SignificanceBisphenol A (BPA), found in many plastic products, has weak estrogenic effects that can be harmful to human health. Thus, structurally related replacements-bisphenol S (BPS) and bisphenol F (BPF)-are coming into wider use with very few data about their biological activities. Here, we compared the effects of BPA, BPS, and BPF on human mammary organoids established from normal breast tissue. BPS disrupted organoid architecture and induced supernumerary branching. At a proteomic level, the bisphenols altered the abundance of common targets and those that were unique to each compound. The latter included proteins linked to tumor-promoting processes. These data highlighted the importance of testing the human health effects of replacements that are structurally related to chemicals of concern.


Benzhydryl Compounds , Carcinogenesis , Estrogens , Mammary Glands, Human , Phenols , Proteome , Sulfones , Benzhydryl Compounds/toxicity , Carcinogenesis/chemically induced , Estrogens/toxicity , Humans , Mammary Glands, Human/drug effects , Mammary Glands, Human/pathology , Organoids/drug effects , Organoids/pathology , Phenols/toxicity , Proteome/drug effects , Proteomics , Sulfones/toxicity
3.
Sci Rep ; 12(1): 317, 2022 01 10.
Article En | MEDLINE | ID: mdl-35013350

This manuscript describes a new method for forming basal-in MCF10A organoids using commercial 384-well ultra-low attachment (ULA) microplates and the development of associated live-cell imaging and automated analysis protocols. The use of a commercial 384-well ULA platform makes this method more broadly accessible than previously reported hanging drop systems and enables in-incubator automated imaging. Therefore, time points can be captured on a more frequent basis to improve tracking of early organoid formation and growth. However, one major challenge of live-cell imaging in multi-well plates is the rapid accumulation of large numbers of images. In this paper, an automated MATLAB script to handle the increased image load is developed. This analysis protocol utilizes morphological image processing to identify cellular structures within each image and quantify their circularity and size. Using this script, time-lapse images of aggregating and non-aggregating culture conditions are analyzed to profile early changes in size and circularity. Moreover, this high-throughput platform is applied to widely screen concentration combinations of Matrigel and epidermal growth factor (EGF) or heparin-binding EGF-like growth factor (HB-EGF) for their impact on organoid formation. These results can serve as a practical resource, guiding future research with basal-in MCF10A organoids.


Cell Culture Techniques, Three Dimensional/instrumentation , Cell Proliferation , High-Throughput Screening Assays , Image Processing, Computer-Assisted , Mammary Glands, Human/physiology , Microscopy, Fluorescence , Organoids , Time-Lapse Imaging , Algorithms , Cell Line , Cell Proliferation/drug effects , Collagen/pharmacology , Drug Combinations , Epidermal Growth Factor/pharmacology , Female , Heparin-binding EGF-like Growth Factor/pharmacology , Humans , Laminin/pharmacology , Mammary Glands, Human/cytology , Mammary Glands, Human/drug effects , Proteoglycans/pharmacology , Time Factors
4.
Nat Commun ; 12(1): 6967, 2021 11 29.
Article En | MEDLINE | ID: mdl-34845227

Breast cancer is now globally the most frequent cancer and leading cause of women's death. Two thirds of breast cancers express the luminal estrogen receptor-positive (ERα + ) phenotype that is initially responsive to antihormonal therapies, but drug resistance emerges. A major barrier to the understanding of the ERα-pathway biology and therapeutic discoveries is the restricted repertoire of luminal ERα + breast cancer models. The ERα + phenotype is not stable in cultured cells for reasons not fully understood. We examine 400 patient-derived breast epithelial and breast cancer explant cultures (PDECs) grown in various three-dimensional matrix scaffolds, finding that ERα is primarily regulated by the matrix stiffness. Matrix stiffness upregulates the ERα signaling via stress-mediated p38 activation and H3K27me3-mediated epigenetic regulation. The finding that the matrix stiffness is a central cue to the ERα phenotype reveals a mechanobiological component in breast tissue hormonal signaling and enables the development of novel therapeutic interventions. Subject terms: ER-positive (ER + ), breast cancer, ex vivo model, preclinical model, PDEC, stiffness, p38 SAPK.


Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Mechanotransduction, Cellular/genetics , Transcriptome , p38 Mitogen-Activated Protein Kinases/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Case-Control Studies , Cell Line, Tumor , Cinnamates/pharmacology , Collagen/chemistry , Collagen/pharmacology , Drug Combinations , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Fulvestrant/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Histones/genetics , Histones/metabolism , Humans , Indazoles/pharmacology , Laminin/chemistry , Laminin/pharmacology , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Phenotype , Proteoglycans/chemistry , Proteoglycans/pharmacology , Tamoxifen/pharmacology , Tissue Culture Techniques , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Reprod Toxicol ; 105: 1-16, 2021 10.
Article En | MEDLINE | ID: mdl-34343637

Mammary gland (MG) undergoes critical points of structural changes throughout a woman's life. During the perinatal and pubertal stages, MG develops through growth and differentiation to establish a pre-mature feature. If pregnancy and lactation occur, the epithelial compartment branches and differentiates to create a specialized structure for milk secretion and nurturing of the newborn. However, the ultimate MG modification consists of a regression process aiming to reestablish the smaller and less energy demanding structure until another production cycle happens. The unraveling of these fascinating physiologic cycles has helped the scientific community elucidate aspects of molecular regulation of proliferative and apoptotic events and remodeling of the stromal compartment. However, greater understanding of the hormonal pathways involved in MG developmental stages led to concern that endocrine disruptors such as bisphenol A (BPA), may influence these specific development/involution stages, called "windows of susceptibility". Since it is used in the manufacture of polycarbonate plastics and epoxy resins, BPA is a ubiquitous chemical present in human everyday life, exerting an estrogenic effect. Thus, descriptions of its deleterious effects on the MG, especially in terms of serum hormone concentrations, hormonal receptor expression, molecular pathways, and epigenetic alterations, have been widely published. Therefore, allied to a didactic description of the main physiological mechanisms involved in different critical points of MG development, the current review provides a summary of key mechanisms by which the endocrine disruptor BPA impacts MG homeostasis at different windows of susceptibility, causing short- and long-term effects.


Benzhydryl Compounds/toxicity , Endocrine Disruptors/toxicity , Mammary Glands, Human/drug effects , Phenols/toxicity , Animals , Female , Homeostasis , Humans , Mammary Glands, Human/growth & development
6.
J Mammary Gland Biol Neoplasia ; 26(3): 297-308, 2021 09.
Article En | MEDLINE | ID: mdl-34463898

Estrogens have pleiotropic effects on many reproductive and non-reproductive tissues and organs including the mammary gland, uterus, ovaries, vagina, and endothelium. Estrogen receptor α functions as the principal mediator of estrogenic action in most of these tissues. Estetrol (E4) is a native fetal estrogen with selective tissue actions that is currently approved for use as the estrogen component in a combined oral contraceptive and is being developed as a menopause hormone therapy (MHT, also known as hormone replacement therapy). However, exogenous hormonal treatments, in particular MHTs, have been shown to promote the growth of preexisting breast cancers and are associated with a variable risk of breast cancer depending on the treatment modality. Therefore, evaluating the safety of E4-based formulations on the breast forms a crucial part of the clinical development process. This review highlights preclinical and clinical studies that have assessed the effects of E4 and E4-progestogen combinations on the mammary gland and breast cancer, focusing in particular on the estrogenic and anti-estrogenic properties of E4. We discuss the potential advantages of E4 over current available estrogen-formulations as a contraceptive and for the treatment of symptoms due to menopause. We also consider the potential of E4 for the treatment of endocrine-resistant breast cancer.


Breast Neoplasms/chemically induced , Contraceptives, Oral, Hormonal/adverse effects , Estetrol/adverse effects , Hormone Replacement Therapy/adverse effects , Mammary Glands, Human/drug effects , Breast Neoplasms/pathology , Female , Humans , Mammary Glands, Human/pathology
7.
Carbohydr Polym ; 269: 118345, 2021 Oct 01.
Article En | MEDLINE | ID: mdl-34294352

This work reports novel chitosan functionalized graphene oxide (GO) nanocomposites combined fluorescence imaging and therapeutic functions in one agent, which can serve as a promising alternative to alleviate related diseases caused hyperinflammation. Briefly, GO was designed to be conjugated with chitosan, fluorescein-labeled peptide, toll-like receptor 4 antibody and hydroxycamptothecin/aloe emodin. We have demonstrated that such nanocomposites could effectively achieve active targeted delivery of pro-apoptotic and anti-inflammatory drugs into inflammatory cells and cause cells apoptosis by acid-responsive drug release. Moreover, confocal fluorescence imaging confirms that the drug-induced inflammatory cells apoptosis could be visualized the light-up fluorescence of fluorescein activated by caspase-3. Meanwhile, inflammatory-related biomarkers have down-regulated after the nanocomposites' treatment in both vitro and vivo experiments consistent with the results in histological sections. In summary, the bifunctional nanocomposites that possess anti-inflammation and fluorescence imaging could serve as a promising therapeutic agent for reducing hyperinflammation caused by numerous diseases.


Anti-Inflammatory Agents/therapeutic use , Apoptosis/physiology , Drug Carriers/chemistry , Inflammation/drug therapy , Nanocomposites/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Antibodies/immunology , Camptothecin/analogs & derivatives , Camptothecin/chemistry , Camptothecin/therapeutic use , Cattle , Cell Line , Chitosan/chemistry , Drug Liberation , Emodin/chemistry , Emodin/therapeutic use , Fluorescent Dyes/chemistry , Graphite/chemistry , Humans , Lipopolysaccharides , Mammary Glands, Human/drug effects , Mammary Glands, Human/pathology , Mastitis/chemically induced , Mastitis/drug therapy , Mastitis/pathology , Mice , Toll-Like Receptor 4/immunology
8.
Molecules ; 26(6)2021 Mar 13.
Article En | MEDLINE | ID: mdl-33805741

A series of novel functionalized methyl 3-(hetero)arylthieno[3,2-b]pyridine-2-carboxylates 2a-2h were synthesized by C-C Pd-catalyzed Suzuki-Miyaura cross-coupling of methyl 3-bromothieno[3,2-b]pyridine-2-carboxylate with (hetero)aryl pinacol boranes, trifluoro potassium boronate salts or boronic acids. Their antitumoral potential was evaluated in two triple negative breast cancer (TNBC) cell lines-MDA-MB-231 and MDA-MB-468, by sulforhodamine B assay. Their effects on the non-tumorigenic MCF-12A cells were also evaluated. The results demonstrated that three compounds caused growth inhibition in both TNBC cell lines, with little or no effect against the non-tumorigenic cells. The most promising compound was further studied concerning possible effects on cell viability (by trypan blue exclusion assay), cell proliferation (by bromodeoxyuridine assay) and cell cycle profile (by flow cytometry). The results demonstrated that the GI50 concentration of compound 2e (13 µM) caused a decreased in MDA-MB-231 cell number, which was correlated with a decreased in the % of proliferating cells. Moreover, this compound increased G0/G1 phase and decreased S phases, when compared to control cells (although was not statistic significant). Interestingly, compound 2e also reduced tumor size using an in ovo CAM (chick chorioallantoic membrane) model. This work highlights the potential antitumor effect of a novel methyl 3-arylthieno[3,2-b]pyridine-2-carboxylate derivative.


Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Thienopyridines/chemical synthesis , Thienopyridines/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Cell Cycle/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chorioallantoic Membrane/surgery , Drug Screening Assays, Antitumor , Female , Humans , Mammary Glands, Human/drug effects , Mammary Glands, Human/pathology , Molecular Structure , Neoplasm Transplantation , Structure-Activity Relationship , Thienopyridines/chemistry , Triple Negative Breast Neoplasms/pathology
9.
J Ethnopharmacol ; 275: 114096, 2021 Jul 15.
Article En | MEDLINE | ID: mdl-33823166

ETHNOPHARMACOLOGICAL RELEVANCE: The increasing use of "kidney"-nourishing Traditional Chinese Medicine (TCM) like Er-xian decoction (EXD) for management of menopausal symptoms and osteoporosis has aroused concerns about their safety, and whether they interact with prescription drugs as both of them act via estrogen receptors (ERs) and regulate serum estradiol. AIM OF THE STUDY: The present study aimed to evaluate whether EXD selectively exerted estrogenic activities and interacted with Selective Estrogen Receptor Modulators (SERMs). MATERIALS AND METHODS: In vivo, mature ovariectomized (OVX) rats were administrated with EXD or combined treatment of EXD and SERMs for 12 weeks. The tissue-selective effect of EXD and its interaction of SERMs were studied in four estrogen sensitive tissues, bone, brain, breast and uterus. In vitro, the interaction of extracts of EXD-treated serum and SERMs in four ER-positive cell lines. RESULTS: In OVX rats, EXD selectively alleviated estrogen deficiency-induced changes in the bone and brain without inducing any estrogenic effects in the breast or uterus. Two-way ANOVA indicated the presence of interactions between EXD and SERMs in OVX rats but EXD did not significantly alter the tissue responses to SERMs in the bone, breast or brain. Indeed, the combined use of EXD and SERMs appeared to suppress the estrogenic effect of raloxifene and tamoxifen in the uterus. Extract of EXD-treated serum directly stimulated cell proliferation or differentiation in human osteosarcoma MG-63, neuroblastoma SHSY5Y, breast cancer MCF-7, and endometrial Ishikawa cells. Two-way ANOVA revealed that EXD-treated serum interacted with SERMs at various concentrations and altered the effects of tamoxifen in MG-63 and MCF-7 cells. CONCLUSIONS: EXD exerted estrogenic effects in a tissue-selective manner and interacted with SERMs. Combined treatment of EXD and SERMs did not hamper the beneficial effects of SERMs on the bone or brain but appeared to moderate the estrogenic effect of SERMs in the uterus.


Drugs, Chinese Herbal/pharmacology , Estrogens/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Alkaline Phosphatase/metabolism , Animals , Body Weight/drug effects , Bone and Bones/drug effects , Bone and Bones/metabolism , Breast/drug effects , Breast/metabolism , Breast/pathology , Cell Line, Tumor , Central Nervous System/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/therapeutic use , Estradiol/pharmacology , Estradiol/therapeutic use , Estrogens/chemistry , Estrogens/therapeutic use , Female , Herb-Drug Interactions/physiology , Hormones/blood , Humans , Mammary Glands, Human/drug effects , Medicine, Chinese Traditional , Models, Biological , Ovariectomy/adverse effects , Raloxifene Hydrochloride/pharmacology , Raloxifene Hydrochloride/therapeutic use , Rats, Sprague-Dawley , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/therapeutic use , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , Uterus/drug effects , Uterus/metabolism , Uterus/pathology , Water
10.
Oncol Rep ; 45(4)2021 04.
Article En | MEDLINE | ID: mdl-33649835

Environmental endocrine disruptor chemicals are substances that can alter the homeostasis of the endocrine system in living organisms. They can be released from several products used in daily activities. Once in the organism, they can disrupt the endocrine function by mimicking or blocking naturally occurring hormones due to their similar chemical structure. This endocrine disruption is the most important cause of the well­known hormone­associate types of cancer. Additionally, it is decisive to determine the susceptibility of each organ to these compounds. Therefore, the present review aimed to summarize the effect of different environmental substances such as bisphenol A, dichlorodiphenyltrichloroethane and polychlorinated biphenyls in both the mammary and the prostate tissues. These organs were chosen due to their association with the hormonal system and their common features in carcinogenic mechanisms. Outcomes derived from the present review may provide evidence that should be considered in future debates regarding the effects of endocrine disruptors on carcinogenesis.


Endocrine Disruptors/pharmacology , Environmental Pollutants/pharmacology , Mammary Glands, Animal/drug effects , Prostate/drug effects , Animals , Benzhydryl Compounds/pharmacology , DDT/pharmacology , Female , Humans , Male , Mammary Glands, Human/drug effects , Phenols/pharmacology , Polychlorinated Biphenyls/pharmacology
11.
Biomed Pharmacother ; 137: 111353, 2021 May.
Article En | MEDLINE | ID: mdl-33578236

BACKGROUND: Mastitis is a common disease occurs in breast-feeding mothers, but published data are poor. This study aimed to study the effects of Tanshinones on treating mastitis. METHODS: Clinical trials performed in 58 breast-feeding mothers were carried out. B-ultrasound and blood test were used to measure the size of breast mass and the change of blood cell counts. BALB/c mice were injected with LPS and then treated by Tanshinone I or Tanshinone IIA/B. Myeloperoxidase (MPO) activity and the release of inflammatory cytokines were tested by MPO kit, RT-qPCR and ELISA. Mouse mammary epithelial cells (mMECs) were isolated and the effects of Tanshinones were measured by conducting CCK-8 assay, flow cytometry, RT-qPCR and ELISA. RESULTS: Patients treated by Cefprozil combined with Tanshinone got better outcomes than patients treated by Cefprozil alone. In animal trials, Tanshinone I and Tanshinone IIA/B significantly reduced MPO activity, and the levels of TNF-α, IL-1ß and IL-6 in serum and mammary gland tissues. In mMECs, Tanshinone I and Tanshinone IIA/B attenuated LPS-induced viability loss and apoptosis. And they effectively inhibited the release of TNF-α, IL-1ß and IL-6. Also, Tanshinone I and Tanshinone IIA/B significantly attenuated LPS-evoked NF-κB activation. CONCLUSION: Tanshinone I and Tanshinone IIA/B have potentials in treating mastitis. The beneficial effects might be through regulating NF-κB activation.


Abietanes/pharmacology , Anti-Infective Agents/pharmacology , Mastitis/drug therapy , NF-kappa B p50 Subunit/metabolism , Abietanes/therapeutic use , Adult , Animals , Anti-Infective Agents/therapeutic use , Apoptosis/drug effects , Breast Feeding/adverse effects , Cephalosporins/pharmacology , Cephalosporins/therapeutic use , Disease Models, Animal , Drug Therapy, Combination , Female , Humans , Inflammation/drug therapy , Interleukin-1beta/drug effects , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/toxicity , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mammary Glands, Human/diagnostic imaging , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Mastitis/chemically induced , Mastitis/metabolism , Mice, Inbred BALB C , Peroxidase/drug effects , Peroxidase/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism , Ultrasonography, Mammary , Cefprozil
12.
Sci Rep ; 11(1): 3761, 2021 02 12.
Article En | MEDLINE | ID: mdl-33580144

Arsenic is reportedly a biphasic inorganic compound for its toxicity and anticancer effects in humans. Recent studies have shown that certain arsenic compounds including arsenic hexoxide (AS4O6; hereafter, AS6) induce programmed cell death and cell cycle arrest in human cancer cells and murine cancer models. However, the mechanisms by which AS6 suppresses cancer cells are incompletely understood. In this study, we report the mechanisms of AS6 through transcriptome analyses. In particular, the cytotoxicity and global gene expression regulation by AS6 were compared in human normal and cancer breast epithelial cells. Using RNA-sequencing and bioinformatics analyses, differentially expressed genes in significantly affected biological pathways in these cell types were validated by real-time quantitative polymerase chain reaction and immunoblotting assays. Our data show markedly differential effects of AS6 on cytotoxicity and gene expression in human mammary epithelial normal cells (HUMEC) and Michigan Cancer Foundation 7 (MCF7), a human mammary epithelial cancer cell line. AS6 selectively arrests cell growth and induces cell death in MCF7 cells without affecting the growth of HUMEC in a dose-dependent manner. AS6 alters the transcription of a large number of genes in MCF7 cells, but much fewer genes in HUMEC. Importantly, we found that the cell proliferation, cell cycle, and DNA repair pathways are significantly suppressed whereas cellular stress response and apoptotic pathways increase in AS6-treated MCF7 cells. Together, we provide the first evidence of differential effects of AS6 on normal and cancerous breast epithelial cells, suggesting that AS6 at moderate concentrations induces cell cycle arrest and apoptosis through modulating genome-wide gene expression, leading to compromised DNA repair and increased genome instability selectively in human breast cancer cells.


Arsenic Trioxide/toxicity , MCF-7 Cells/drug effects , Mammary Glands, Human/drug effects , Apoptosis/drug effects , Arsenic/metabolism , Arsenic Trioxide/metabolism , Arsenic Trioxide/pharmacology , Arsenicals/pharmacology , Breast Neoplasms/genetics , Cell Cycle/drug effects , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Female , Gene Expression/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mammary Glands, Human/metabolism , Primary Cell Culture , Tumor Cells, Cultured
13.
Int J Mol Sci ; 22(2)2021 Jan 12.
Article En | MEDLINE | ID: mdl-33445675

Mycotoxins are present in everyday diet as common food and feed pollutants. A part of them is still concerned as so-called emerging mycotoxins. Due to the lack of toxicity data, the safety limits and detail molecular mechanism have been not established yet for all of them. Alternariol (AOH), as one of these mycotoxins, produced by Alternaria species, is so far reported as an estrogenic, genotoxic, and immunomodulatory agent; however, its direct effect on human health is not known. Especially, in the case of hormone-dependent tissues which are sensitive to both endogenic, as well as external estrogenic agents, it might be crucial to assess the effect of AOH. Thus, this study evaluated how exposure to AOH affects viability and motility of the human normal mammary gland epithelial in vitro model. We observed that AOH significantly affects viability of cells in a time- and dose-dependent manner. Moreover, the induction of oxidative stress, DNA damage, and cell cycle arrest in the G2/M cell cycle phase was observed. The motility of 184A1 cells was also significantly affected. On the molecular level, AOH induced antioxidative stress response via activation of Nuclear factor erythroid 2-related factor 2 (NRF2) signaling pathway agents, as well as decrease in the phosphorylation of protein kinase B (Akt) and p44/42 (ERK 1-2) molecules, indicating that AOH might affect crucial signaling pathways in both physiological and pathophysiological processes in breast tissue.


Breast/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Epithelial Cells/drug effects , Lactones/pharmacology , Mammary Glands, Human/drug effects , Mycotoxins/pharmacology , Alternaria/metabolism , Breast/metabolism , Cell Cycle Checkpoints/drug effects , Cell Division/drug effects , Cell Line , DNA Damage/drug effects , Epithelial Cells/metabolism , Female , G2 Phase/drug effects , Humans , Mammary Glands, Human/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Phosphorylation/drug effects , Signal Transduction/drug effects
14.
Int Immunopharmacol ; 91: 107324, 2021 Feb.
Article En | MEDLINE | ID: mdl-33385711

It is well-established that lysine-specific demethylase 1 (LSD1) is the first identified histone demethylase. Based on its demethylase enzymatic activity, LSD1 plays a pivotal role in vast range of cellular processes and cancers, but the understanding of its effects on inflammation is relatively limited. Using in vivo models of lipopolysaccharide (LPS)-induced inflammation and in vitro assays in mouse mammary epithelial cells, we identified the novel regulatory roles and underlying mechanisms of LSD1 on LPS-induced mastitis. Mammary gland and cells were collected for the following experiments after treatment. Histological changes were determined by H&E. Western blot analysis was used to detect the protein expression. ELISA and real-time PCR were used to evaluate protein and mRNA expression of inflammatory genes. Our results showed that LPS treatment resulted in a significant increase in LSD1 protein expression. GSK-LSD1 is a selective inhibitor of LSD1 enzyme activity. Treatment of mice with GSK-LSD1 inhibited LSD1 activity, reduced inflammatory cells recruitment to tissues and attenuated LPS-induced damage in mammary gland. Mechanistic investigations suggested that LSD1 inhibition led to the increase of histone H3K4me2 and H3K9me2. Furthermore, GSK-LSD1 inhibition of LSD1 further inhibited nuclear factor κ-B (NF-κB) signaling cascades, and subsequently inhibited the production of cytokines (TNF-α, IL-6 and IL-1ß) in mammary gland. Taken together, our data reveal LSD1 as a potential regulator of inflammation and improve our understanding of epigenetic control on inflammation.


Epigenesis, Genetic , Epithelial Cells/enzymology , Histone Demethylases/metabolism , Mammary Glands, Human/enzymology , Mastitis/enzymology , Animals , Anti-Inflammatory Agents/pharmacology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/genetics , Humans , Inflammation Mediators/metabolism , Lipopolysaccharides , Mammary Glands, Human/drug effects , Mammary Glands, Human/pathology , Mastitis/chemically induced , Mastitis/genetics , Mastitis/prevention & control , Mice, Inbred BALB C , NF-kappa B/metabolism
15.
J Ethnopharmacol ; 271: 113863, 2021 May 10.
Article En | MEDLINE | ID: mdl-33485972

ETHNOPHARAMCOLOGICAL RELEVANCE: The age-long folkloric use of Uvaria chamae roots in the management of nipple discharge that is not related to pregnancy, childbirth or nursing but as a result of excessive production of prolactin (hyperprolactinemia) is yet to be substantiated with scientific data. AIM OF THE STUDY: This study investigated the anti-hyperprolactinemic activities of aqueous extract of Uvaria chamae roots (AEUCR) and associated biochemical changes in chlorpromazine (CPZ)-induced hyperprolactinemic female Wistar rats. MATERIALS AND METHODS: A total of sixty female rats (207.40 ± 2.69 g) were assigned into 6 groups: A-F. Animals in Group A received 0.5 ml of distilled water only whilst the 7 days CPZ-treated female rats (to induce hyperprolactinemia) in groups B, C, D, E, and F also received distilled water, 2.5 mg/kg body weight of bromocriptine (reference drug), 0.71, 1.41 2.83 mg/kg body weight of AEUCR for 28 days. RESULTS: AEUCR contained a total of 15 (75%) amino acids with seven (46.67%) being essential amino acids and eight (53.33%) as non-essential amino acids. Administration of CPZ increased (p < 0.05) the levels of prolactin and testosterone, and reduced (p < 0.05) the levels of estradiol, progesterone, follicle stimulating hormone (FSH), luteinizing hormone (LH), dopamine, triiodothyronine (T3) and tetraiodothyroxine (T4). Chlorpromazine also increased the levels of serum urea, creatinine, total protein, albumin, globulin, bilirubin, alanine aminotransferase (ALT), aspartate aminotransferase (AST) and alkaline phosphatase (ALP) of the animals. In contrast, AEUCR significantly (p < 0.05) reduced the CPZ-induced increases in the levels of prolactin and testosterone, and increased the levels of CPZ-induced reduction in the progesterone, estradiol, FSH, LH, dopamine, T3 and T4. The AEUCR also reversed (p < 0.05) the CPZ-induced related increases in the levels of urea, creatinine, total protein, albumin, globulin, bilirubin, ALT, AST and ALP similar to the trends in the distilled water- and bromocriptine-treated controls. The CPZ-induced remarkable increase in the size of lactating alveolus and lactiferous duct distribution in the mammary gland were restored to normal tubule-alveolar female pattern mammary glands, composed of branching ducts and small alveoli budding off the ducts. CONCLUSION: The study concluded that aqueous extract of Uvaria chamae root exhibited anti-hyperprolactinemic activity by restoring prolactin and dopamine levels and tubule-alveolar female pattern in female rats. It also ameliorated CPZ-induced changes in the liver and kidney function indices. This study justifies the folkloric use of Uvaria chamae root in the management of abnormal discharge by the nipples that is unrelated to pregnancy, childbirth and nursing.


Hyperprolactinemia/drug therapy , Plant Extracts/therapeutic use , Plant Roots/chemistry , Uvaria/chemistry , Animals , Chlorpromazine/toxicity , Estradiol/metabolism , Female , Follicle Stimulating Hormone/metabolism , Humans , Hyperprolactinemia/chemically induced , Kidney/drug effects , Liver/drug effects , Luteinizing Hormone/metabolism , Mammary Glands, Human/drug effects , Mammary Glands, Human/pathology , Medicine, Traditional , Plant Extracts/pharmacology , Progesterone/metabolism , Prolactin/metabolism , Rats, Wistar , Testosterone/metabolism , Thyroid Hormones/metabolism , Water/chemistry
16.
Sci Rep ; 10(1): 15188, 2020 09 16.
Article En | MEDLINE | ID: mdl-32938954

Small molecule inhibitors targeting BCL2 are explored as anticancer therapeutics. Previously, we have reported identification and characterization of a novel BCL2 inhibitor, Disarib. Disarib induced cancer cell death in a BCL2 dependent manner in different cancer cell lines and mouse tumor models when it was administered intraperitoneally. In the present study, using two syngeneic mouse models, breast adenocarcinoma (EAC) and Dalton's lymphoma (DLA), we show that oral administration of Disarib resulted in significant tumor regression in a concentration dependent manner. Importantly, tumor developed in both female and male mice were equally sensitive to Disarib. Further, we have investigated the toxicity of Disarib in normal cells. Single dose toxicity analysis of Disarib in male and female mice after oral administration revealed no significant variations compared to control group for parameters such as body weight, food and water consumption and behavioural changes which were analysed for the entire period of study. Haematological and histopathological analyses also did not show any significant difference from the control groups. Thus, our results reveal safe use of Disarib as a small molecule inhibitor and provide the foundation for investigation of other preclinical studies.


Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Drug-Related Side Effects and Adverse Reactions/diagnosis , Indoles/therapeutic use , Lymphoma/drug therapy , Mammary Glands, Human/drug effects , Mammary Neoplasms, Experimental/drug therapy , Thiadiazoles/therapeutic use , Administration, Oral , Animals , Blood Cell Count , Body Weight/drug effects , Cell Line, Tumor , Female , Hematopoiesis/drug effects , Humans , Indoles/pharmacology , Male , Mammary Glands, Human/pathology , Mammary Neoplasms, Experimental/diagnosis , Mice , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Thiadiazoles/pharmacology
17.
Horm Mol Biol Clin Investig ; 41(3)2020 Jul 31.
Article En | MEDLINE | ID: mdl-32735552

Background The impact of hormones on the development of breast cancer is despite extensive studies, incompletely understood. Combined estrogen-progestogen treatment augments the risk for breast cancer beyond that of estrogen alone, according to numerous studies. The role of breast cell proliferation as a promoter in the development and growth of breast cancer is well recognized. Materials and methods Seventy-nine patients from three randomised trials were subject to a re-analysis of breast cell proliferation: (1) 22 women received continuous combined treatment with oral estradiol (E2) 2 mg/norethisterone acetate (NETA) 1 mg once daily for 3 months. (2) Thirty-seven women received 2 months of sequential treatment with oral conjugated equine estrogens (CEE) 0.625 mg daily combined with medroxyprogesterone acetate (MPA) 5 mg for 14/28 days of each cycle. (3) Twenty women received oral estradiol-valerate (E2V) 2 mg daily combined with levonorgestrel (LNG) intrauterine system (IUS), 20 µg/24 h for 2 months. Fine needle aspiration (FNA) (studies 1 and 3) and core needle biopsy (CNB) (study 2) were used for the assessment of breast cell proliferation. Results There were no baseline proliferation differences, but at the end of treatment there was a highly significant between-group difference for E2V/LNG IUS versus the other two groups (p = 0.0025). E2/NETA and CEE treatments gave a 4-7-old increase in proliferation during treatment (p = 0.04) and (p = 0.007), respectively, which was absent in the E2V/LNG group, showing a significant correlation with insulin-like growth factor binding protein-3 (IGFBP-3) serum levels. Conclusion E2V in combination with very low serum concentrations of LNG in the IUS gives no increase in proliferation in the normal breast.


Breast Neoplasms/etiology , Estrogen Replacement Therapy/adverse effects , Levonorgestrel/adverse effects , Mammary Glands, Human/drug effects , Progestins/adverse effects , Administration, Oral , Aged , Cell Proliferation , Female , Humans , Levonorgestrel/administration & dosage , Levonorgestrel/therapeutic use , Mammary Glands, Human/pathology , Medroxyprogesterone Acetate/administration & dosage , Medroxyprogesterone Acetate/adverse effects , Medroxyprogesterone Acetate/therapeutic use , Middle Aged , Norethindrone Acetate/administration & dosage , Norethindrone Acetate/adverse effects , Norethindrone Acetate/therapeutic use , Progestins/administration & dosage , Progestins/therapeutic use , Uterus
18.
Mol Cell Endocrinol ; 518: 110927, 2020 12 01.
Article En | MEDLINE | ID: mdl-32645345

Breast cancer risk from pesticides may be missed if effects on mammary gland are not assessed in toxicology studies required for registration. Using US EPA's registration documents, we identified pesticides that cause mammary tumors or alter development, and evaluated how those findings were considered in risk assessment. Of 28 pesticides that produced mammary tumors, EPA's risk assessment acknowledges those tumors for nine and dismisses the remaining cases. For five pesticides that alter mammary gland development, the implications for lactation and cancer risk are not assessed. Many of the mammary-active pesticides activate pathways related to endocrine disruption: altering steroid synthesis in H295R cells, activating nuclear receptors, or affecting xenobiotic metabolizing enzymes. Clearer guidelines based on breast cancer biology would strengthen assessment of mammary gland effects, including sensitive histology and hormone measures. Potential cancer risks from several common pesticides should be re-evaluated, including: malathion, triclopyr, atrazine, propylene oxide, and 3-iodo-2-propynyl butylcarbamate (IPBC).


Mammary Glands, Animal/drug effects , Mammary Glands, Human/drug effects , Pesticides/toxicity , Practice Guidelines as Topic/standards , Toxicity Tests/standards , Animals , Breast Neoplasms/chemically induced , Breast Neoplasms/pathology , Endocrine Disruptors/pharmacology , Female , Government Regulation , Humans , Risk Assessment , Toxicity Tests/methods , United States , United States Environmental Protection Agency/legislation & jurisprudence , United States Environmental Protection Agency/standards
19.
Medicine (Baltimore) ; 99(28): e20727, 2020 Jul 10.
Article En | MEDLINE | ID: mdl-32664067

RATIONALE: Most gastric cancer patients are diagnosed at mid- to late-stage and lose the chance of radical surgery, medical treatment is especially important to prolong the survival of patients. Apatinib mesylate, which is a small molecule vascular endothelial growth factor receptor 2 tyrosine kinase inhibitor, could be used as antiangiogenesis therapy for gastric cancer. PATIENT CONCERNS: A 67-year-old man sought medical care for upper abdominal discomfort. DIAGNOSIS: The patient was diagnosed as mixed medullary differentiated gastric adenocarcinoma, and immunohistochemistry suggested HER-2 (2+). INTERVENTIONS: The patient received chemotherapy consisting of oxaliplatin combined with S-1 as first-line treatment, and targeted therapy with apatinib mesylate as second-line treatment. OUTCOMES: After 4 months of first-line chemotherapy, the patient received apatinib treatment immediately at a dose of 500 mg/d orally and died of cardiac arrest with 8.5 months of overall survival. During this period of targeted therapy with apatinib mesylate, this male patient suffered mammary gland development besides other common adverse reactions. LESSONS: This case report is the first to report the case of male mammary gland development after oral apatinib.


Antineoplastic Agents/adverse effects , Carcinoma, Medullary/drug therapy , Mammary Glands, Human/drug effects , Pyridines/adverse effects , Stomach Neoplasms/drug therapy , Aged , Humans , Male
20.
Mol Biol Rep ; 47(7): 4923-4932, 2020 Jul.
Article En | MEDLINE | ID: mdl-32557190

Experimental evidence highlights the importance of dietetic factors on breast cancer. In this work we aimed to analyze the effects two oils, corn oil (rich in n-6 polyunsaturated fatty acids -PUFA-) and extra virgin olive oil (EVOO), on oxidative stress in an animal model of breast carcinogenesis. Female rats were fed a low-fat control, a high-corn oil, or a high-EVOO diet from weaning or after induction with 7,12-dimethylbenz[a]anthracene at 53 days. Animals were euthanized at 36, 51, 100 and 246 days of age. We analyzed antioxidant enzymes (mRNA and activity of superoxide dismutase, glutathione peroxidase and catalase), non-enzymatic capacity (oxidized and reduced glutathione) and DNA damage (8-oxo-dG) in tumors and mammary gland at different ages. We also analyzed lipid peroxidation (isoprostanes in serum and lipofuscin in liver). Results indicated a decrease in the enzymatic antioxidant capacity and increased oxidative stress in mammary gland of healthy young animals after a short period of high-fat diets intake, followed by an adaptation to chronic dietary intervention. After induction both diets, especially the one high in n-6 PUFA, increased the oxidized glutathione. In tumors no clear effects of the high-fat diets were observed, although in the long-term lipofuscin and 8-oxo-dG suggested greater oxidative damage by effect of the n-6 PUFA-rich diet. Considering the differential effects of these diets on mammary carcinogenesis that we have previously reported, this study suggests that these high-fat diets could have an effect on oxidative stress that would lead to different signaling pathways.


Corn Oil/pharmacology , Diet , Mammary Neoplasms, Experimental/metabolism , Olive Oil/pharmacology , Oxidative Stress , Animals , Corn Oil/administration & dosage , DNA Damage , Female , Glutathione/metabolism , Humans , Isoprostanes/blood , Lipofuscin/metabolism , Liver/drug effects , Liver/metabolism , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Olive Oil/administration & dosage , Rats , Rats, Sprague-Dawley
...