Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Biomed Pharmacother ; 155: 113762, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36182733

ABSTRACT

Current targeted- and immuno-therapies have prolonged the overall survival of non-small cell lung cancer (NSCLC) patients by few months in a small percentage of patients responding to these treatments. This situation has prompted us to investigate the anticancer potential of the Origanum majorana Essential Oil (OMEO). In this pre-clinical study and using two major human NSCLC, namely A549 and LNM35, we demonstrated that OMEO significantly decreases the viability of these cells and the growth of their pre-formed colonies in vitro in a concentration-dependent manner and partly via the induction of caspase 3/7-dependent cell death and downregulation of survivin. Moreover, OMEO significantly slow down the growth of A549 and LNM35 tumor xenografts in the CAM and in nude mice models in vivo. Furthermore, OMEO significantly reduces in vitro A549 and LNM35 cancer cell migration and invasion, and the incidence and growth of lymph nodes metastasis in vivo in nude mice xenografted subcutaneously with the highly metastatic LNM35 cells. Three months of treatment of mice with OMEO did not affect blood, kidney, and liver functions. Our study demonstrates that OMEO is a safe and robust anticancer option.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Oils, Volatile , Origanum , Humans , Mice , Animals , Lung Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Survivin , Mice, Nude , Oils, Volatile/pharmacology , Oils, Volatile/therapeutic use , Caspase 3/metabolism , Cell Movement , Cell Line, Tumor , Cell Proliferation
2.
Nutrients ; 14(16)2022 Aug 15.
Article in English | MEDLINE | ID: mdl-36014840

ABSTRACT

Junctional epithelia are common sites for pathological transformations. In mice, the stratified epithelium of the forestomach joins the simple glandular epithelium of the cardia at the limiting ridge. We previously demonstrated the expression of vitamin A receptors in the gastric stem/progenitor cells and their progeny and found that excess retinoic acid enhances cellular dynamics of gastric epithelium. This study examines how deficiency of vitamin A would alter gastric epithelial stem cell lineages. Three-week-old mice of both genders were weaned and fed with a vitamin A deficient (VAD) diet for 4 or 8 months. Sex- and weight-matched littermate mice received a standard (control) diet. To label S-phase cells, all mice received a single intraperitoneal injection of 5-bromo-2-deoxyuridine before being euthanized. Stomach tissues were processed for microscopic examination and protein analysis to investigate stem cell lineages using different stains, lectins, or antibodies. The Student's t-test was used to compare quantified data showing differences between control and VAD groups. Eight-month-vitamin-A deficiency caused enlarged forestomach and overgrowth of the squamocolumnar junction with metaplastic and dysplastic cardiac glands, formation of intramucosal cysts, loss of surface mucosal integrity, increased amount of luminal surface mucus, and upregulation of trefoil factor 1 and H+,K+-ATPase. These changes were associated with decreased cell proliferation and upregulation of p63. In conclusion, vitamin A is necessary for maintaining gastric epithelial integrity and its deficiency predisposes the mouse stomach to precancerous lesions.


Subject(s)
Stomach , Vitamin A , Animals , Cell Lineage , Epithelium/metabolism , Female , Gastric Mucosa/metabolism , Humans , Male , Mice , Vitamin A/metabolism
3.
BMC Psychiatry ; 22(1): 209, 2022 03 21.
Article in English | MEDLINE | ID: mdl-35313855

ABSTRACT

BACKGROUND: To date, only few studies have investigated ghrelin levels in bipolar disorders, and all have exclusively measured acylated ghrelin, with none investigating total ghrelin (acylated and des-acylated). We aimed to investigate peripheral levels of acylated and total ghrelin in subjects experiencing a manic episode of bipolar disorder. METHODS: Peripheral levels of acylated and total ghrelin were measured in hospitalised medicated individuals recovering from a manic episode. Enzyme-linked immunosorbent assays (ELISA) were used to measure ghrelin levels in patients and compared with healthy controls. The relationship between ghrelin levels in bipolar disorder, self-reported hunger measures, demographic and clinical parameters was investigated with correlational analyses. RESULTS: Twenty-four subjects (15 males, 9 females) recovering from mania and 27 matched healthy controls (13 males, 14 females) were recruited for the study. Mean values of both acylated (187 vs.520 pg/mL) and total ghrelin (396 vs. 648 pg/mL) were significantly reduced in bipolar disorder (p = 0.001). Ghrelin levels correlated positively with markers of illness severity and negatively with prescribed mood stabilizers, second-generation antipsychotics, weight and body mass index. CONCLUSION: Peripheral measurements of acylated and total ghrelin were both reduced in bipolar disorder patients compared to healthy controls. Whilst illness severity promotes higher ghrelin levels, pharmacological treatment and weight gain exercise the opposite effect.


Subject(s)
Antipsychotic Agents , Bipolar Disorder , Bipolar Disorder/drug therapy , Body Mass Index , Female , Ghrelin , Humans , Male , Mania
4.
Biology (Basel) ; 10(7)2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34203400

ABSTRACT

Patients with gastric mucosal erosions are predisposed to chronic gastritis, ulcer or even cancer. The repair of mucosal erosions involves several events including proliferation of gastric epithelial stem cells. The aim of this study was to investigate the effects of the probiotic mixture of De Simone Formulation on gastric epithelial stem cell lineages in mouse models of gastric mucosal erosions. Gastric erosions were induced by a single oral gavage of 80% ethanol containing 15 mg/mL acetylsalicylic acid (5 mL/kg) following a daily dose of probiotic mixture (5 mg/day/mouse) for 10 days. In another protocol, erosions were induced by a daily gavage of acetylsalicylic acid (400 mg/kg/day/mouse) for 5 days before or after daily administration of probiotic mixture for 5 days. Control mice received water gavage for 10 days. All mice were injected with bromodeoxyuridine two hours before sacrifice to label S-phase cells. The stomachs of all mice were processed for histological examination, lectin binding, and immunohistochemical analysis. The results reveal that mice that received probiotics before or after the induction of erosion showed a decrease in erosion index with an increase in gastric epithelial stem/progenitor cell proliferation and enhanced production of mucus, trefoil factors, and ghrelin by mucous and enteroendocrine cell lineages. These mice also showed restoration of the amount of H+,K+-ATPase and pepsinogen involved in the production of the harsh acidic environment by parietal and chief cell lineages. In conclusion, this study demonstrates the beneficial effects of probiotics against gastric mucosal erosion and highlights the involvement and modulation of proliferative stem cells and their multiple glandular epithelial cell lineages.

5.
Life (Basel) ; 10(8)2020 Aug 12.
Article in English | MEDLINE | ID: mdl-32806566

ABSTRACT

Stem cells have attracted many scientists because of their unique properties and therapeutic applications. However, very little is known on the environmental toxins that could affect their biological features. This study focuses on the consequences of the exposure of a cell line representative of the mouse gastric stem/progenitor (mGS) cells to diesel exhaust particles (DEPs). These immortal cells were cultured using routine protocols. The DEPs were added to the culture media at 1, 10, and 100 µg/mL for 1 to 72 h. The cells were assayed for their viability, migration, oxidative stress, and the expression of genes specific for cell proliferation, pluripotency, and death. DEPs induced a reduction in the metabolic activity of mGS cells, only at a high concentration of 100 µg/mL. However, no significant effects were detected on cell migration, oxidative stress markers (glutathione and thiobarbituric acid reactive substances), and cell death related proteins/genes. Interestingly, these findings were associated with down-regulation of Notch 2 and 3 and Bmi-1 proteins and activation of STAT3 involved in the regulation of the fate of stem cells. In conclusion, this study demonstrates that mGS cells have some resistance to oxidative stress and apoptosis when exposed to DEPs at the expense of their stemness.

6.
Asian-Australas J Anim Sci ; 33(2): 349-359, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31010974

ABSTRACT

Orchastric changes in the mammary glands are vital, especially during the lactaction. The secretary epithelial cells together with the supporting myoepithelial and stromal cells function cordially to secrete milk.Increase in the number of luminal epithelial cells and a decrease in adipocytes are visible during lactation, whereas the reverse happens in the involution. However, an early involution occurs if the epithelial transdifferente towards adipocytes in lactation period. We aimed to inhibit the adipocyte transdifferentiation of luminal cells by restraining the PPARγ pathway. Linolenic acid (LA) and thiazolidinediones (TZDs) induced adipogenesis in mammary epithelial cells were conducted in monolayer, mixed culture as well as in transwell plate co-culture with mammary myoepithelial cells.Co-culture with myoepithelial cells showed higher adipogenic gene expression in epithelial cells under LA+TZDs treatment. Increase in the expressions of PPARγ, C/EBPα and vimentin in both mRNA as well as protein levels were observed.Whereas,BADGE treatment blocked LA+TZDs induced adipogenesis, as it could not show a significant rise in adipose related markers. Although comparative results were found in both mixed culture and monolayer conditions, co-culture technic found to work better than the others. In summary, antagonizing PPARγ pathway in the presence of myoepithelial cells can significantly reduce the adipogenisis in epithelial cells, suggestingtherapeutic inhibition of PPARγ can be considered to counter early involution or excessive adipogenesis in mammary epithelium in animals.

7.
Int J Mol Sci ; 20(16)2019 Aug 10.
Article in English | MEDLINE | ID: mdl-31405107

ABSTRACT

Probiotics are used in the management of some gastrointestinal diseases. However, little is known about their effects on normal gastric epithelial biology. The aim of this study was to explore how the probiotic mixture VSL#3 affects gastric cell lineages in mice with a special focus on protective and aggressive factors. Weight-matching littermate male mice (n = 14) were divided into treated and control pairs. The treated mice received VSL#3 (5 mg/day/mouse) by gastric gavage for 10 days. Control mice received only the vehicle. Food consumption and bodyweight were monitored. All mice were injected intraperitoneally with bromodeoxyuridine (120 mg/Kg bodyweight) two hours before sacrificed to label S-phase cells. Stomach tissues were processed for lectin- and immunohistochemical examination. ImageJ software was used to quantify immunolabeled gastric epithelial cells. Real-time quantitative polymerase chain reaction was used to provide relative changes in expression of gastric cell lineages specific genes. Results revealed that treated mice acquired (i) increased production of mucus, trefoil factor (TFF) 1 and TFF2, (ii) decreased production of pepsinogen, and (iii) increased ghrelin-secreting cells. No significant changes were observed in bodyweight, food consumption, cell proliferation, or parietal cells. Therefore, VSL#3 administration amplifies specific cell types specialized in the protection of the gastric epithelium.


Subject(s)
Gastric Mucosa/metabolism , Pepsinogen A/genetics , Probiotics/pharmacology , Trefoil Factors/genetics , Animals , Down-Regulation , Gastric Mucosa/cytology , Gastric Mucosa/microbiology , Gastric Mucosa/ultrastructure , Male , Mice , Mice, Inbred C57BL , Probiotics/administration & dosage , Up-Regulation
8.
Int Immunopharmacol ; 40: 79-89, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27584057

ABSTRACT

Inflammation is set off when innate immune cells detect infection or tissue injury. Tight control of the severity, duration, and location of inflammation is an absolute requirement for an appropriate balance between clearance of injured tissue and pathogens versus damage to host cells. Impeding the risk associated with the imbalance in the inflammatory response requires precise identification of potential therapeutic targets involved in provoking the inflammation. Toll-like receptors (TLRs) primarily known for the pathogen recognition and subsequent immune responses are being investigated for their pathogenic role in various chronic diseases. A mammalian homologue of Drosophila Toll receptor 4 (TLR4) was shown to induce the expression of genes involved in inflammatory responses. Signaling pathways via TLR4 activate various transcription factors like Nuclear factor kappa-light-chain-enhancer (NF-κB), activator protein 1 (AP1), Signal Transducers and Activators of Transcription family of transcription factors (STAT1) and Interferon regulatory factors (IRF's), which are the key players regulating the inflammatory response. Inhibition of these targets and their upstream signaling molecules provides a potential therapeutic approach to treat inflammatory diseases. Here we review the therapeutic targets involved in TLR-4 signaling pathways that are critical for suppressing chronic inflammatory disorders.


Subject(s)
Inflammation/metabolism , Toll-Like Receptor 4/metabolism , Animals , Humans , Inflammation/drug therapy , Interferon Regulatory Factors/metabolism , NF-kappa B/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...