Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 193
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Cell Rep ; 38(7): 110363, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35172147

ABSTRACT

Thymic atrophy reduces naive T cell production and contributes to increased susceptibility to viral infection with age. Expression of tissue-restricted antigen (TRA) genes also declines with age and has been thought to increase autoimmune disease susceptibility. We find that diminished expression of a model TRA gene in aged thymic stromal cells correlates with impaired clonal deletion of cognate T cells recognizing an autoantigen involved in atherosclerosis. Clonal deletion in the polyclonal thymocyte population is also perturbed. Distinct age-associated defects in the generation of antigen-specific T cells include a conspicuous decline in generation of T cells recognizing an immunodominant influenza epitope. Increased catalase activity delays thymic atrophy, and here, we show that it mitigates declining production of influenza-specific T cells and their frequency in lung after infection, but does not reverse declines in TRA expression or efficient negative selection. These results reveal important considerations for strategies to restore thymic function.


Subject(s)
Aging/immunology , Antigens/immunology , Immunity , Self Tolerance/immunology , T-Lymphocytes/immunology , Animals , Antioxidants/pharmacology , Apolipoproteins B/metabolism , Atrophy , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Catalase/metabolism , Dietary Supplements , Immunity/drug effects , Immunodominant Epitopes/immunology , Mice, Inbred C57BL , Mice, Transgenic , Orthomyxoviridae/drug effects , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/immunology , Oxidation-Reduction , Oxidative Stress/drug effects , Self Tolerance/drug effects , Stromal Cells/drug effects , Stromal Cells/enzymology , T-Lymphocytes/drug effects , Thymus Gland/pathology
2.
Life Sci Alliance ; 4(12)2021 12.
Article in English | MEDLINE | ID: mdl-34593556

ABSTRACT

Worldwide, ∼196 million are afflicted with endometriosis, a painful disease in which endometrial tissue implants and proliferates on abdominal peritoneal surfaces. Theories on the origin of endometriosis remained inconclusive. Whereas up to 90% of women experience retrograde menstruation, only 10% develop endometriosis, suggesting that factors that alter peritoneal environment might contribute to endometriosis. Herein, we report that whereas some gut bacteria promote endometriosis, others protect against endometriosis by fermenting fiber to produce short-chain fatty acids. Specifically, we found that altered gut microbiota drives endometriotic lesion growth and feces from mice with endometriosis contained less of short-chain fatty acid and n-butyrate than feces from mice without endometriosis. Treatment with n-butyrate reduced growth of both mouse endometriotic lesions and human endometriotic lesions in a pre-clinical mouse model. Mechanistic studies revealed that n-butyrate inhibited human endometriotic cell survival and lesion growth through G-protein-coupled receptors, histone deacetylases, and a GTPase activating protein, RAP1GAP. Our findings will enable future studies aimed at developing diagnostic tests, gut bacteria metabolites and treatment strategies, dietary supplements, n-butyrate analogs, or probiotics for endometriosis.


Subject(s)
Bacteria/metabolism , Butyrates/administration & dosage , Butyrates/metabolism , Endometriosis/metabolism , Endometriosis/microbiology , Gastrointestinal Microbiome , Protective Agents/administration & dosage , Protective Agents/metabolism , Signal Transduction/drug effects , Animals , Cell Line, Transformed , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Endometriosis/drug therapy , Endometriosis/pathology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Feces/chemistry , Feces/microbiology , Female , Heterografts , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Shelterin Complex/metabolism , Signal Transduction/genetics , Stromal Cells/drug effects , Stromal Cells/metabolism , Telomere-Binding Proteins/metabolism , Transfection
3.
Histol Histopathol ; 36(8): 833-844, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33829420

ABSTRACT

Cells cultured as monolayers proliferate well, but do not sustain their differentiation characteristics. Previous studies have investigated the interactions between cells and growth factors or cytokines by establishing either in vivo or in vitro three-dimensional (3D) cultures. Using porcine uterine epithelial cells and endometrial cells, the current study was designed to develop a 3D uterine culture system and investigate the response to hormone treatment. Formation of the 3D uterine model was similar to that of uterus from the group supplemented with calcium and magnesium, and the addition of these ions altered the spectrum of basement membrane degrading enzyme expression and activity. In particular, the epithelial cell junctions in the 3D model most closely resembled those of an actual uterus when the medium was supplemented with calcium and magnesium; the intercellular basement membrane structure was also tall under these conditions. The study confirmed that Casp-3 expression was lowest in the P4 (progesterone) treatment group, and this hormone was the most potent stimulus for formation of the endometrial cell layer. Therefore, the addition of calcium and magnesium plays an important role in the formation of a 3D uterine model, and the addition of P4 hormone mimics uterine thickening by stimulating growth of the epithelial cell layer.


Subject(s)
Endometrium/cytology , Endometrium/pathology , Estradiol/pharmacology , Progesterone/pharmacology , Stromal Cells/cytology , Animals , Coculture Techniques , Endometrium/metabolism , Female , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism , Swine , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism , Vascular Endothelial Growth Factor A/metabolism
4.
Toxicol In Vitro ; 71: 105067, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33301902

ABSTRACT

In response to the need for reliable cellular models that reflect complex tumor microenvironmental properties, and enable more precise testing of anti-cancer therapeutics effects on humans, a co-culture platform for in-vitro model that enhances the physiology of breast cancer (BC) microenvironment is presented. A six well imaging plate wherein each macro-well contains several separate compartments was designed. Three-dimensional (3D) cancer spheroids are generated and cultured in the inner compartment which is embossed with an array of nano-liter micro-chambers made of hydrogel. Stromal cells are cultured in the outer chambers. The two cell types are cultured side-by-side, sharing a common space, thus enabling extra-cellular communication via secreted molecules. As proof of concept, a model of BC tumor microenvironment was recapitulated by co-cultivating 3D MCF7 spheroids in the presence of tumor-associated macrophages (TAMs). The presence of TAMs induced an aggressive phenotype by promoting spheroid growth, enhancing survivin expression levels and enabling invasive behavior. Moreover, TAMs influenced the response of BC spheroids to cytotoxic treatment as well as hormonal drug therapy, and enhanced the effects of nitric oxide donor. The platform enables time-lapse imaging and treatment without losing spatial location of the measured spheroids, thereby allowing measurements and analysis at individual-object resolution in an easy and efficient manner.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Evaluation, Preclinical/methods , Breast Neoplasms/drug therapy , Coculture Techniques , Doxorubicin/pharmacology , Humans , Hydrogels , MCF-7 Cells , Macrophages/drug effects , Models, Biological , Spheroids, Cellular/drug effects , Stromal Cells/drug effects , Tamoxifen/pharmacology , Triazenes/pharmacology , Tumor Microenvironment , U937 Cells
5.
Sci Rep ; 10(1): 11404, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32647159

ABSTRACT

There is currently no therapy to limit the development of cardiac fibrosis and consequent heart failure. We have recently shown that cardiac fibrosis post-myocardial infarction (MI) can be regulated by resident cardiac cells with a fibrogenic signature and identified by the expression of PW1 (Peg3). Here we identify αV-integrin (CD51) as an essential regulator of cardiac PW1+ cells fibrogenic behavior. We used transcriptomic and proteomic approaches to identify specific cell-surface markers for cardiac PW1+ cells and found that αV-integrin (CD51) was expressed in almost all cardiac PW1+ cells (93% ± 1%), predominantly as the αVß1 complex. αV-integrin is a subunit member of the integrin family of cell adhesion receptors and was found to activate complex of latent transforming growth factor beta (TGFß at the surface of cardiac PW1+ cells. Pharmacological inhibition of αV-integrin reduced the profibrotic action of cardiac PW1+CD51+ cells and was associated with improved cardiac function and animal survival following MI coupled with a reduced infarct size and fibrotic lesion. These data identify a targetable pathway that regulates cardiac fibrosis in response to an ischemic injury and demonstrate that pharmacological inhibition of αV-integrin could reduce pathological outcomes following cardiac ischemia.


Subject(s)
Integrin alphaV/drug effects , Myocardial Infarction/drug therapy , Snake Venoms/therapeutic use , Stromal Cells/drug effects , Animals , Cells, Cultured , Drug Evaluation, Preclinical , Fibrosis , Integrin alphaV/physiology , Kruppel-Like Transcription Factors/analysis , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardium/pathology , Myocytes, Cardiac/metabolism , RNA, Messenger/biosynthesis , Single-Cell Analysis , Snake Venoms/pharmacology , Stromal Cells/chemistry , Transforming Growth Factor beta1/pharmacology
6.
Molecules ; 25(6)2020 Mar 13.
Article in English | MEDLINE | ID: mdl-32183239

ABSTRACT

There is increasing interest in the potential of natural compounds to treat diseases, such as endometriosis, a gynecological disorder that affects 10-15% of women of reproductive age, and it is related to severe pelvic pain and infertility. We have evaluated the in vitro effects of rutin and the aqueous bark, roots, and leaf extracts (ABE, ARE, and ALE, respectively) and isolated components of Uncaria guianensis on stromal cells from eutopic endometrium and lesions of patients with endometriosis. Two- and three-dimensional cultures were used to assess the cell death and production of reactive oxygen species (ROS), cytokines and growth factors of cells following exposure to these natural products. The applied treatments did not reduce cellular viability, but ROS production did increase. In addition, significant increases in the levels of interleukin (IL)-15, IL-17A, IL-4, IL-6, tumor necrosis factor-α, and vascular endothelium growth factor were observed when 2D-cells from endometrium of patients with endometriosis were treated with ABE, while exposure to ALE induced significant increases in epidermal growth factor in lesion cells.


Subject(s)
Endometriosis/pathology , Plant Extracts/pharmacology , Rutin/pharmacology , Uncaria/chemistry , Alkaloids/chemistry , Biomarkers/metabolism , Cell Survival/drug effects , Cells, Cultured , Cytokines/metabolism , Female , Fluorescence , Humans , Inflammation Mediators/metabolism , Phenols/chemistry , Reactive Oxygen Species/metabolism , Spheroids, Cellular/drug effects , Stromal Cells/drug effects , Stromal Cells/metabolism
7.
J Reprod Immunol ; 139: 103104, 2020 06.
Article in English | MEDLINE | ID: mdl-32172005

ABSTRACT

OBJECTIVES: To evaluate the anti-inflammatory and anti-angiogenetic effects of Tokishakuyakusan (TSS), a traditional Japanese medicine (Kampo), and its ingredients, ferulic acid (FA) and paeoniflorin (PA) on endometriotic stromal cells (ESC) and peritoneal macrophages. STUDY DESIGN: Endometriotic tissues were obtained from 16 patients and peritoneal macrophages were obtained from 11 patients that had undergone laparoscopic surgery for ovarian endometriosis. ESC isolated from endometriotic tissues and peritoneal macrophages were cultured, and pre-treated with 300 µg/mL of TSS, 500 µM FA or 50 µM PA. ESC and peritoneal macrophages were then stimulated with IL-1ß. Concentrations of IL-8 and VEGF protein in supernatants were then detected and measured using specific ELISAs. TSS (4 g/kg body weight) was orally administered to female Sprague-Dawley rats. The concentration of FA in plasma and uteri was measured using liquid chromatography-mass spectrometry with tandem mass spectrometry (LC-MS/MS).  RESULTS: TSS and FA but not PA decreased the secretion of inflammatory cytokine (IL-8) and angiogenic factor (VEGF) in ESC. TSS and FA also suppressed the secretion of inflammatory cytokine (IL-8) from peritoneal macrophages. FA was detected in plasma and in uterine tissues after the oral administration of TSS to rats. CONCLUSIONS: Our study demonstrates that TSS has anti-inflammatory and anti-angiogenic effects on endometriosis related cells by controlling inflammatory cytokine and growth factor secretion from cells, and these effects, at least partially, may be due to the direct effects of the TSS ingredient FA.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Coumaric Acids/pharmacology , Drugs, Chinese Herbal/pharmacology , Endometriosis/therapy , Endometrium/pathology , Glucosides/pharmacology , Macrophages, Peritoneal/immunology , Monoterpenes/pharmacology , Stromal Cells/immunology , Adult , Angiogenesis Inhibitors , Animals , Cytokines/metabolism , Female , Humans , Inflammation Mediators/metabolism , Macrophages, Peritoneal/drug effects , Medicine, Kampo , Middle Aged , Stromal Cells/drug effects
8.
Nutrients ; 12(2)2020 Jan 30.
Article in English | MEDLINE | ID: mdl-32019160

ABSTRACT

Vitamin D status has been implicated in obesity and adipose tissue inflammation. In the present study, we explored the effects of dietary vitamin D supplementation on adipose tissue inflammation and immune cell population, and the effects of in vitro 1,25-dihydroxyvitamin D3 (1,25-(OH)2D3) treatment on pro-inflammatory cytokine production by stromal vascular cells (SVCs) and adipocytes in lean and high-fat diet-induced obese mice. The results show that epididymal fat Mcp-1 and Rantes mRNA levels, which were higher in obese mice compared with lean mice, were significantly down-regulated by vitamin D supplementation. While obese mice had higher numbers of macrophages and natural killer (NK) cells within adipose tissue, these remained unaltered by vitamin D supplementation. In accordance with these in vivo findings, the in vitro 1,25(OH)2D3 treatment decreased IL-6, MCP-1, and IL-1ß production by SVCs from obese mice, but not by adipocytes. In addition, 1,25(OH)2D3 treatment significantly decreased Tlr2 expression and increased mRNA levels of Iκba and Dusp1 in SVCs. These findings suggest that vitamin D supplementation attenuates inflammatory response in adipose tissue, especially in SVCs, possibly through inhibiting NF-κB and MAPK signaling pathways in SVCs but not by the inhibition of macrophage infiltration.


Subject(s)
Adipocytes/drug effects , Calcitriol/pharmacology , Obesity/immunology , Stromal Cells/drug effects , Vitamins/pharmacology , Adipocytes/immunology , Adipose Tissue/drug effects , Adipose Tissue/immunology , Animals , Cytokines/drug effects , Cytokines/immunology , Diet, High-Fat , Disease Models, Animal , Inflammation , Mice , Mice, Obese , Obesity/therapy , Stromal Cells/immunology
9.
Reproduction ; 159(4): 453-463, 2020 04.
Article in English | MEDLINE | ID: mdl-31990677

ABSTRACT

The differentiation of endometrial stromal cells (ESC), named decidualization, is essential to regulate trophoblast invasion and to support pregnancy establishment and progression. Decidualization follows ESC proliferation and it has been described that cell cycle arrest contributes to a proper decidualization. Interestingly, resveratrol, a natural compound derived from grapes with antioxidant properties, has been widely studied in relation to endometrial health. However, little is known about the effect of resveratrol supplementation during decidualization. Therefore, in this study we evaluate the effect of resveratrol supplementation during decidualization. We used primary and immortalized human ESC and we decidualized them in vitro with a decidualization cocktail containing medroxyprogesterone acetate, estradiol and 8-Bromo-cyclic AMP. Pre-decidualized cells were further treated with the decidualization cocktail supplemented with resveratrol. Our results show that resveratrol supplementation increased, in a dose-dependent manner, the expression levels of prolactin and IGFBP1 (RT-PCR and ELISA), indicating an enhanced in vitro decidualization of human ESC. This enhanced decidualization was accompanied by a decrease in cell proliferation (crystal violet and CellTiter proliferation assay) and by changes in the mRNA levels of key cell cycle regulators (RT-PCR). Furthermore, resveratrol supplementation seemed to enhance decidualization by reinforcing the effect of the decidualization cocktail. We believe that resveratrol could to be an effective supplementation to reinforce hormone action during human ESC decidualization and that further insights into resveratrol action and its interaction with estradiol and progesterone signaling pathways could facilitate the identification of new therapeutic strategies for the improvement of women's health.


Subject(s)
Antioxidants/pharmacology , Decidua/drug effects , Resveratrol/pharmacology , Adult , Antioxidants/therapeutic use , Cell Cycle Proteins/metabolism , Cell Line , Decidua/cytology , Decidua/metabolism , Dietary Supplements , Drug Evaluation, Preclinical , Female , Humans , Menstruation Disturbances/therapy , Primary Cell Culture , Resveratrol/therapeutic use , Stromal Cells/drug effects
10.
J Orthop Res ; 38(2): 297-310, 2020 02.
Article in English | MEDLINE | ID: mdl-31471919

ABSTRACT

Giant cell tumor of bone (GCTB) is a locally aggressive destructive bone lesion. The management of pulmonary metastasis and local recurrence after the surgical treatment of GCTB remains a challenge. Pathologically, stromal cells in GCTB are known as primary neoplastic cells and are recognized as incompletely differentiated preosteoblasts. Therefore, inducing GCTB stromal cells to differentiate into cells with a mature osteoblastic phenotype may stop tumor growth and recurrence. In this study, we aimed to investigate how simvastatin, a clinically approved and commonly used statin that has been known to promote the maturation of cells of the osteogenic lineage, affects GCTB stromal cells. We found that simvastatin effectively inhibited cell viability by suppressing proliferation and by inducing apoptosis in GCTB stromal cells. Moreover, simvastatin treatment upregulated the expression of genes related to osteogenic maturation, such as runt-related transcription factor 2, osteopontin, and osteocalcin, and increased the mineralization of the extracellular matrix in GCTB stromal cells. Ingenuity pathway analysis was used to discover that the vitamin D receptor pathway was involved in the simvastatin-induced osteogenic differentiation of GCTB stromal cells by upregulating the 1,25-dihydroxyvitamin D metabolism. Taken together, this in vitro study demonstrates the antitumor and differentiation-promoting effects of simvastatin on GCTB stromal cells and suggests the possibility of using simvastatin as an adjuvant therapy for GCTB. These findings support further clinical investigation of the efficacy of using simvastatin as an adjuvant therapy for GCTB to reduce recurrence and distant metastasis after surgical treatment. © 2019 Orthopedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:297-310, 2020.


Subject(s)
Giant Cell Tumor of Bone/drug therapy , Hypolipidemic Agents/therapeutic use , Simvastatin/therapeutic use , Cell Differentiation/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Giant Cell Tumor of Bone/metabolism , Humans , Hypolipidemic Agents/pharmacology , Simvastatin/pharmacology , Stromal Cells/drug effects , Stromal Cells/metabolism , Vitamin D/analogs & derivatives , Vitamin D/metabolism
11.
Fertil Steril ; 112(5): 947-958.e3, 2019 11.
Article in English | MEDLINE | ID: mdl-31371049

ABSTRACT

OBJECTIVE: To investigate whether phytoestrogens (genistein and daidzein) alter in vitro decidualization of human endometrial stromal cells (ESCs). DESIGN: Isolated primary ESCs were exposed to phytoestrogens and decidualized in vitro. SETTING: Academic fertility center. PATIENT(S): Twenty fertile oocyte donors attending the IVI Valencia clinic. INTERVENTION(S): Treatment of ESC with phytoestrogens at 0, 10, 20, 50, and 100 µM. MAIN OUTCOME MEASURE(S): The ESC proliferation was analyzed by MTS assay. In vitro decidualization was induced in the presence of phytoestrogens by medroxyprogesterone acetate/cyclic adenosine 3':5' monophosphate and evaluated by prolactin (PRL) ELISA and F-actin immunostaining. The Ki67 proliferative marker was analyzed by immunofluorescence. The ESC apoptosis was assessed by annexin V/propidium iodide detection using flow cytometry. Estrogen (ERß) and P receptor (PR) localization were evaluated by immunofluorescence. RESULT(S): The ESC exposed to 0, 19, 20, 50, and 100 µM of genistein, daidzein, and genistein + daidzein showed a dose-dependent proliferation decrease. After 48-96 hours of culture, this reduction was significant in the presence of 50 µM of phytoestrogens versus 10 µM untreated ESC. The ESC decidualized in the presence of phytoestrogens did not rearrange their cytoskeletons and showed a significant decrease in PRL secretion compared with untreated decidualized ESCs (dESCs). However, phytoestrogens did not alter proliferative status or the percentage of viable/apoptotic cells in dESC compared with untreated dESC. During decidualization, phytoestrogens induced the same nuclear translocation of ERß and PR as the control dESC. CONCLUSION(S): This study reveals that high doses of phytoestrogens could affect the in vitro decidualization process.


Subject(s)
Endometrium/drug effects , Genistein/pharmacology , Isoflavones/pharmacology , Phytoestrogens/pharmacology , Stromal Cells/drug effects , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Decidua/cytology , Decidua/drug effects , Decidua/physiology , Dose-Response Relationship, Drug , Endometrium/cytology , Endometrium/physiology , Female , Humans , Signal Transduction/drug effects , Signal Transduction/physiology , Stromal Cells/physiology
12.
PLoS One ; 14(5): e0217365, 2019.
Article in English | MEDLINE | ID: mdl-31136609

ABSTRACT

Plastic polymers can be combined with additives that modify physical properties and stability of the material. However, the biocompatibility of those additives is not well known. The objective of the study was to characterize the impact of zinc stearate-a common additive-through the development of a novel three-dimensional (3-D) in vitro platform with endometrial cells from domestic cats. Epithelial and stromal cells from adult uteri were isolated and cultured in medium supplemented with 3% Matrigel for two weeks in plastic tissue culture dishes that had been identified as polystyrene with and without zinc stearate by Raman, FTIR, and X-ray fluorescence spectroscopies. Three-dimensional cell structures that were obtained were measured and categorized by shape. Cell viability, proliferation, differentiation, organization, and apoptosis then were assessed by immuno-staining. Results indicated that zinc stearate did not affect 3-D endometrial cell structure morphology, viability, or cellular composition. This first study of a new in vitro platform will be useful for studies testing the influence of other additives, drugs, or exogenous hormones.


Subject(s)
Cell Culture Techniques/methods , Endometrium/cytology , Plastics/toxicity , Animals , Apoptosis/drug effects , Cats , Cell Culture Techniques/instrumentation , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/drug effects , Female , Organoids/cytology , Organoids/drug effects , Polystyrenes/toxicity , Stearic Acids/toxicity , Stromal Cells/cytology , Stromal Cells/drug effects
13.
Biomed Pharmacother ; 116: 108972, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31103825

ABSTRACT

Insulin resistance (IR) is one of the characteristic features of equine metabolic syndrome (EMS). Presently, the only therapies of choice are caloric restrictions combined with mineral supplementation, which might improve insulin sensitivity. In this study we investigated the effect of Haematococcus pluvialis algae water extract enriched in bioaccumulation process in magnesium ions (Hp_Mg(II)) on equine adipose derived mesenchymal stromal stem cells, in which insulin resistance was induced by palmitic acid (IR-EqASCs). For this purpose, chemical characterization of H. pluvialis was performed with special emphasis on the analysis of minerals composition, total phenolic and carotenoids contents, as well as scavenging activity. To examine the influence of H. pluvialis extract on IR-EqASCs, various methods of molecular biology and microscopic observations (i.e., immunofluorescence staining, SEM, gene expression by RT-qPCR, proliferative and metabolic cells activity analysis) were applied to investigate in vitro viability, oxidative stress markers and apoptosis-related factor accumulation, along with insulin resistance-related genes expression. Obtained results show, that Hp_Mg(II) significantly improves proliferative and metabolic activity of IR-EqASCs, shortens their population doubling time, improves their clonogenic potential and reduces expression of apoptosis related genes. Moreover, anti-oxidative effect of extract was presented.


Subject(s)
Adipose Tissue/cytology , Insulin Resistance , Magnesium/pharmacology , Microalgae/chemistry , Animals , Antioxidants/pharmacology , Cell Proliferation/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Horses , Insulin Resistance/genetics , Ions , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Oxidation-Reduction , Palmitic Acid , Stromal Cells/drug effects , Stromal Cells/metabolism , Water
14.
Phytother Res ; 33(4): 1044-1054, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30838714

ABSTRACT

Resveratrol, a phytoalexin polyphenol, has antiproliferative, antiangiogenic, anti-inflammatory, and antioxidant properties. The present study has assessed the effect of resveratrol treatment on the expression of insulin-like growth factor-1 (IGF-1) and hepatocyte growth factor (HGF) in endometrial stromal cells (ESCs) from women with and without endometriosis. Endometrial tissues were obtained from 40 endometriotic patients and 15 nonendometriotic control women. After the enzymatic digestion, 13 eutopic ESCs (EuESCs), 8 ectopic ESCs (EESCs), and 11 control ESCs (CESCs) were treated with resveratrol (100 µM) for 6, 24, and 48 hr. The gene and protein expressions of IGF-1 and HGF were measured using real-time polymerase chain reaction and enzyme-linked immunosorbent assay methods, respectively. Results showed that resveratrol treatment decreased significantly the gene expression of IGF-1 and HGF in EuESCs, EESCs, and CESCs (p < 0.05). The effect of resveratrol treatment on the reduction of IGF-1 gene expression was statistically more noticeable in EESCs compared with CESCs (p < 0.05). Also, in the case of HGF gene expression, the reducing effect of resveratrol treatment was statistically more considerable in EESCs compared with EuESCs and CESCs (p < 0.05 and p < 0.01, respectively). The IGF-1 and HGF protein production decreased significantly in EuESCs and EESCs (p < 0.05) but not in CESCs. These findings suggest that resveratrol treatment could reduce the expression of IGF-1 and HGF in ESCs especially in EESCs, which play a pivotal role in disease progression.


Subject(s)
Endometriosis/pathology , Endometrium/drug effects , Endometrium/pathology , Hepatocyte Growth Factor/genetics , Insulin-Like Growth Factor I/genetics , Peritoneal Diseases/pathology , Resveratrol/pharmacology , Adult , Case-Control Studies , Cells, Cultured , Endometriosis/genetics , Endometriosis/metabolism , Endometrium/metabolism , Female , Gene Expression/drug effects , Hepatocyte Growth Factor/metabolism , Humans , Insulin-Like Growth Factor I/metabolism , Middle Aged , Peritoneal Diseases/genetics , Peritoneal Diseases/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism , Young Adult
15.
J Cell Physiol ; 234(5): 6298-6312, 2019 05.
Article in English | MEDLINE | ID: mdl-30259980

ABSTRACT

Endometriosis is a chronic gynecological inflammatory disorder in which immune system dysregulation is thought to play a role in its initiation and progression. Due to altered sex steroid receptor concentrations and other signaling defects, eutopic endometriotic tissues have an attenuated response to progesterone. This progesterone-resistance contributes to lesion survival, proliferation, pain, and infertility. The current agency-approved hormonal therapies, including synthetic progestins, GnRH agonists, and danazol are often of limited efficacy and counterproductive to fertility and cause systemic side effects due to suppression of endogenous steroid hormone levels. In the current study, we examined the effects of curcumin (CUR, diferuloylmethane), which has long been used as an anti-inflammatory folk medicine in Asian countries for this condition. The basal levels of proinflammatory and proangiogenic chemokines and cytokines expression were higher in primary cultures of stromal cells derived from eutopic endometrium of endometriosis (EESC) subjects compared with normal endometrial stromal cells (NESC). The treatment of EESC and NESC with CUR significantly and dose-dependently reduced chemokine and cytokine secretion over the time course. Notably, CUR treatment significantly decreased phosphorylation of the IKKα/ß, NF-κB, STAT3, and JNK signaling pathways under these experimental conditions. Taken together, our findings suggest that CUR has therapeutic potential to abrogate aberrant activation of chemokines and cytokines, and IKKα/ß, NF-κB, STAT3, and JNK signaling pathways to reduce inflammation associated with endometriosis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Curcumin/pharmacology , Endometriosis/pathology , Endometrium/drug effects , Signal Transduction/drug effects , Stromal Cells/drug effects , Cytokines/drug effects , Cytokines/immunology , Cytokines/metabolism , Endometriosis/immunology , Endometriosis/metabolism , Endometrium/immunology , Endometrium/pathology , Female , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , NF-kappa B/drug effects , NF-kappa B/metabolism , Stromal Cells/immunology , Stromal Cells/pathology
16.
J Pharmacol Exp Ther ; 368(3): 490-502, 2019 03.
Article in English | MEDLINE | ID: mdl-30591528

ABSTRACT

The combination of decellularized nerve allograft and adipose-derived stromal cells (ASCs) represents a good alternative to nerve autograft for bridging peripheral nerve defects by providing physical guidance and biologic cues. However, the regeneration outcome of acellular nerve allograft (ANA) is often inferior to autograft. Therefore, we hypothesized that acetyl-l-carnitine (ALCAR) treatment and implantation of ASC-embedded ANA would work synergistically to promote nerve regeneration. Seventy rats were randomly allocated into seven experimental groups (n = 10), including the healthy control group, sham surgery group, autograft group, ANA group, ANA + ASCs group, ANA + ALCAR group (50 mg/kg for 2 weeks), and ANA + ASCs + ALCAR (50 mg/kg for 2 weeks) group. All grafts were implanted to bridge long-gap (10-mm) sciatic nerve defects. Functional, electrophysiological, and morphologic analysis was conducted during the experimental period. We found that ALCAR potentiated the survival and retention of transplanted ASCs and upregulated the expression of neurotrophic factor mRNAs in transplanted grafts. Sixteen weeks following implantation in the rat, the ANA supplemented by ASCs was capable of supporting reinnervation across a 10-mm sciatic nerve gap, with results close to that of the autografts in terms of functional, electrophysiological, and histologic assessments. Results demonstrated that ALCAR treatment improved regenerative effects of ANA combined with ASCs on reconstruction of a 10-mm sciatic nerve defect in rat comparable to those of autograft.


Subject(s)
Acetylcarnitine/administration & dosage , Adipose Tissue/transplantation , Allografts/transplantation , Nerve Regeneration/physiology , Sciatic Neuropathy/therapy , Stromal Cells/transplantation , Acellular Dermis/drug effects , Adipose Tissue/drug effects , Adipose Tissue/physiology , Allografts/drug effects , Allografts/physiology , Animals , Male , Nerve Regeneration/drug effects , Random Allocation , Rats , Rats, Wistar , Sciatic Neuropathy/drug therapy , Sciatic Neuropathy/pathology , Stromal Cells/drug effects , Stromal Cells/pathology , Vitamin B Complex/administration & dosage
17.
Nutrients ; 10(7)2018 Jul 13.
Article in English | MEDLINE | ID: mdl-30011861

ABSTRACT

Vitamin D (VD) deficiency is associated with reproductive failure. However, the relationship between VD and maternal immunity remains unclear. We investigated the clinical efficacy of VD in maternal T-helper (Th) cytokines in 276 infertile women and examined for Th1 and Th2 cells based on the deficient, insufficient, and sufficient serum 25-hydroxyvitamin D3 (25[OH]VD) levels (<12, 12⁻30, and >30 ng/mL, respectively). Most infertile women had a low-level of VD (87.3%). Immunological tests of pre-/post-VD supplementation were performed in patients who were deficient and insufficient in VD. Of 23 patients, 11 (47.8%) exhibited sufficient VD levels after supplementation. Th1/Th2 cell ratio in patients with insufficient VD was significantly decreased after supplementation (p = 0.004). After supplementation, serum 25(OH)VD levels of the patients: 11 in the sufficient group showed significant decreases in Th1 cell level and Th1/Th2 cell ratio (p = 0.032 and 0.010, respectively), whereas no significant differences in Th1/Th2 cell ratio were recognized in the insufficient group. Furthermore, mid-luteal endometrial biopsies (n = 18) were processed for primary cultures and measured interferon [IFN]-γ and interleukin [IL]-4 in condition media. Decidualizing cultures with 1,25-dihydroxvitamin D3 (1,25[OH]2VD) decreased IFN-γ. Sufficient VD supplementation in women with insufficient VD may optimize maternal T-helper cytokines during pregnancy via rebalancing the Th1/Th2 cell ratio.


Subject(s)
Calcifediol/deficiency , Cholecalciferol/administration & dosage , Cytokines/metabolism , Dietary Supplements , Endometrium/drug effects , Infertility, Female/drug therapy , Th1 Cells/drug effects , Th2 Cells/drug effects , Vitamin D Deficiency/drug therapy , Adult , Biomarkers/blood , Calcifediol/blood , Cells, Cultured , Cholecalciferol/adverse effects , Cytokines/immunology , Dietary Supplements/adverse effects , Endometrium/immunology , Endometrium/metabolism , Female , Humans , Infertility, Female/blood , Infertility, Female/diagnosis , Infertility, Female/immunology , Primary Cell Culture , Prospective Studies , Stromal Cells/drug effects , Stromal Cells/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th1-Th2 Balance/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism , Time Factors , Treatment Outcome , Vitamin D Deficiency/blood , Vitamin D Deficiency/diagnosis , Vitamin D Deficiency/immunology
18.
Cell Mol Biol (Noisy-le-grand) ; 64(6): 58-64, 2018 May 15.
Article in English | MEDLINE | ID: mdl-29808802

ABSTRACT

To examine the exact role of flavored Guilu Erxian decoction, a Traditional Chinese Medicine (TCM) in the treatment of cisplatin-induced side-effects in bone marrow mesenchymal stem cells (BM-MSCs). BM-MSCs were isolated from bone marrow collected from SD rats and identified by flow cytometry. Cells were cultivated in MEM alpha medium containing 5% (TCM-L), 10% (TCM-M) and 20% (TCM-H) dosages of flavored Guilu Erxian decoction with or without cisplatin. Cell viability was determined through CCK-8 and thymidine analog 5-ethynyl-2'-deoxyuridine (EdU) staining assay. Flow cytometry was used to determine cell cycle and apoptosis. The expression of p21 and cleaved-caspase-3 were examined using Western blot assay. The PI3K-AKT-mTOR pathway associated proteins, including p-PI3K, p-AKT and p-mTOR, were also examined by Western blot assay. CCK-8 and EdU staining assay demonstrated that cisplatin could inhibit cell proliferation in BM-MSCs in a dose and time dependent manner. Further, cisplatin could induce apoptosis through increasing G0/G1 cell cycle arrest, p21 and cleaved-caspase-3 expression. However, these phenomena would be significantly alleviated when adding the serum containing flavored Guilu Erxian decoction. Furthermore, the PI3K-AKT-mTOR pathway activation could be inhibited by cisplatin in BM-MSCs, while flavored Guilu Erxian decoction treatment successfully abrogated this effect. Combination of flavored Guilu Erxian decoction and cisplatin could reduce the damage to BM-MSCs. This indicates that the flavored Guilu Erxian decoction can enhance the possibility of BM-MSCs repairing and rehabilitating the normal function of injured tissues induced by cisplatin, which could provide a new direction for therapeutic applications.


Subject(s)
Bone Marrow Cells/drug effects , Cisplatin/toxicity , Drugs, Chinese Herbal/pharmacology , Mesenchymal Stem Cells/drug effects , Animals , Apoptosis/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Caspase 3/biosynthesis , Caspase 3/genetics , Cell Cycle/drug effects , Cell Division/drug effects , Cell Separation , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation/drug effects , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Random Allocation , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology
19.
Br J Cancer ; 119(2): 164-169, 2018 07.
Article in English | MEDLINE | ID: mdl-29755119

ABSTRACT

BACKGROUND: The tumour-stroma ratio (TSR) has proven to be an independent prognostic factor in colon cancer. METHODS: Haematoxylin eosin tissue slides of patients from the AVANT trial were microscopically scored for TSR and categorised as stroma -low or stroma -high. Scores were correlated to the primary and secondary endpoint disease-free survival (DFS) and overall survival (OS). RESULTS: Patients with stroma-high tumours (N = 339, 28%) had a significantly shorter DFS (p < 0.001) compared to stroma-low tumours (N = 824, 68%). In the bevacizumab-FOLFOX-4 arm, DFS was significantly shorter compared to FOLFOX-4 in stroma-low tumours, with a hazard ratio (HR) of 1.94 (95% CI 1.24-3.04; p = 0.004). In stroma-high tumours a trend for better DFS was seen in bevacizumab-FOLFOX-4 vs. FOLFOX-4 (HR 0.61 (95% CI 0.35-1.07; p = 0.08)). For bevacizumab-XELOX vs. FOLFOX-4, this was not seen (stroma-low HR 1.07 (95% CI 0.64-1.77; p = 0.80); stroma-high HR 0.78 (95% CI 0.47-1.30; p = 0.35)). OS showed the same pattern for bevacizumab-FOLFOX-4 vs. FOLFOX-4 with a HR of 2.53 (95% CI 1.36-4.71; p = 0.003) for stroma-low and HR 0.50 (95% CI 0.22-1.14; p = 0.10) for stroma-high tumours. For bevacizumab-XELOX vs. FOLFOX-4, HR 1.13 (95% CI 0.55-2.31; p = 0.74) for stroma-low tumours and HR 0.74 (95% CI 0.37-1.51; p = 0.41) for stroma-high tumours. CONCLUSIONS: This exploratory analysis suggests a significantly shorter DFS and OS in stroma-low tumours with addition of bevacizumab to intravenous oxaliplatin-based chemotherapy, contrary to stroma-high tumours, where a beneficial trend is observed.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Colonic Neoplasms/drug therapy , Prognosis , Stromal Cells/drug effects , Aged , Aged, 80 and over , Bevacizumab/administration & dosage , Capecitabine , Colonic Neoplasms/pathology , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Disease-Free Survival , Female , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Humans , Leucovorin/administration & dosage , Male , Middle Aged , Neoplasm Staging , Organoplatinum Compounds/administration & dosage , Oxaloacetates , Stromal Cells/pathology
20.
Fertil Steril ; 109(4): 728-734.e2, 2018 04.
Article in English | MEDLINE | ID: mdl-29397924

ABSTRACT

OBJECTIVE: To investigate the impact of the androgen precursor dehydroepiandrosterone (DHEA) on the decidualization of human endometrial stromal cells isolated from women of advanced reproductive age. DESIGN: In vitro study. SETTING: University research institute. PATIENT(S): Proliferative phase primary human endometrial stromal fibroblasts (hESFs) were isolated from women of advanced reproductive age (n = 16; mean age, 44.7 ± 2.3). None of the women were receiving hormone therapy or had endometriosis. INTERVENTION(S): Isolated hESFs were decidualized in vitro by incubation with P (1 µM) and cAMP (0.1 mg/mL) in the presence, or absence, of DHEA (10 nM, 100 nM). MAIN OUTCOME MEASURE(S): Secretion of androgens was assessed by ELISA. Expression of decidualization markers and endometrial receptivity markers was assessed by quantitative polymerase chain reaction and ELISA. RESULT(S): Decidualization responses were retained in hESF isolated from women of advanced reproductive age. Supplementation with DHEA increased androgen biosynthesis and concentrations of T and dihydrotestosterone were ∼3× greater after coincubation with DHEA compared with hESF stimulated with decidualization alone. Addition of DHEA to decidualized hESF increased expression of the decidualization markers IGFBP1 and PRL and the endometrial receptivity marker SPP1. DHEA enhanced secretion of IGFBP1, PRL, and SPP1 proteins maximally by day 8 of the decidualization time course concomitant with peak androgen concentrations. CONCLUSION(S): These novel results demonstrate DHEA can enhance in vitro decidualization responses of hESF from women of advanced reproductive age. Supplementation with DHEA during the receptive phase may augment endometrial function and improve pregnancy rates in natural or assisted reproductive cycles.


Subject(s)
Cell Proliferation/drug effects , Decidua/drug effects , Dehydroepiandrosterone/pharmacology , Fibroblasts/drug effects , Maternal Age , Reproductive Health , Stromal Cells/drug effects , Adult , Biomarkers/metabolism , Cells, Cultured , Decidua/cytology , Decidua/metabolism , Dihydrotestosterone/metabolism , Female , Fibroblasts/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 1/metabolism , Middle Aged , Osteopontin/metabolism , Prolactin/metabolism , Stromal Cells/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL