13 results on '"Borahay, Mostafa A."'
Search Results
2. Simvastatin Inhibits Wnt/β-Catenin Pathway in Uterine Leiomyoma.
- Author
-
El Sabeh M, Saha SK, Afrin S, and Borahay MA
- Subjects
- Adolescent, Adult, Cell Proliferation drug effects, Cell Proliferation genetics, Cells, Cultured, Clinical Trials, Phase II as Topic, Double-Blind Method, Down-Regulation drug effects, Down-Regulation genetics, Female, Humans, Leiomyoma genetics, Leiomyoma metabolism, Middle Aged, Primary Cell Culture, Randomized Controlled Trials as Topic, Uterine Neoplasms genetics, Uterine Neoplasms metabolism, Young Adult, beta Catenin antagonists & inhibitors, beta Catenin metabolism, Leiomyoma pathology, Simvastatin pharmacology, Uterine Neoplasms pathology, Wnt Signaling Pathway drug effects
- Abstract
The Wnt/β-catenin pathway is upregulated in uterine leiomyomas, the most common benign tumors in the female reproductive tract. Simvastatin is an antihyperlipidemic drug, and previous in vitro and in vivo reports showed that it may have therapeutic effects in treating leiomyomas. The objective of this study was to examine the effects of simvastatin on the Wnt/β-catenin signaling pathway in leiomyoma. We treated primary and immortalized human leiomyoma cells with simvastatin and examined its effects using quantitative real-time polymerase chain reaction, Western blotting, and immunocytochemistry. We also examined the effects using human leiomyoma tissues from an ongoing randomized controlled trial in which women with symptomatic leiomyoma received simvastatin (40 mg) or placebo for 3 months prior to their surgery. The results of this study revealed that simvastatin significantly reduced the expression of Wnt4 and its co-receptor LRP5. After simvastatin treatment, levels of total β-catenin and its active form, nonphosphorylated β-catenin, were reduced in both cell types. Additionally, simvastatin reduced the expression of Wnt4 and total β-catenin, as well as nonphosphorylated β-catenin protein expression in response to estrogen and progesterone. Simvastatin also inhibited the expression of c-Myc, a downstream target of the Wnt/β-catenin pathway. The effect of simvastatin on nonphosphorylated-β-catenin, the key regulator of the Wnt/β-catenin pathway, was recapitulated in human leiomyoma tissue. These results suggest that simvastatin may have a beneficial effect on uterine leiomyoma through suppressing the overactive Wnt/β-catenin pathway., (© The Author(s) 2021. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
3. Simvastatin modulates estrogen signaling in uterine leiomyoma via regulating receptor palmitoylation, trafficking and degradation.
- Author
-
Afrin S, El Sabeh M, Islam MS, Miyashita-Ishiwata M, Malik M, Catherino WH, Akimzhanov AM, Boehning D, Yang Q, Al-Hendy A, Segars JH, and Borahay MA
- Subjects
- Adolescent, Adult, Animals, Cell Line, Tumor, Cell Survival, Double-Blind Method, Female, Humans, Hydroxymethylglutaryl-CoA Reductase Inhibitors therapeutic use, Leiomyoma drug therapy, Lipoylation, Mice, Middle Aged, Protein Transport, Proteolysis, Signal Transduction drug effects, Simvastatin therapeutic use, Uterine Neoplasms drug therapy, Young Adult, Estrogen Receptor alpha metabolism, Estrogens metabolism, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Leiomyoma metabolism, Simvastatin pharmacology, Uterine Neoplasms metabolism
- Abstract
Uterine leiomyomas or fibroids are the most common tumors of the female reproductive tract. Estrogen (E
2 ), a steroid-derived hormone, and its receptors (ERs), particularly ER-α, are important drivers for the development and growth of leiomyomas. We previously demonstrated that simvastatin, a drug used for hyperlipidemia, also possesses anti-leiomyoma properties. The aim of this work is to investigate the impact of simvastatin on ER-α signaling in leiomyoma cells, including its expression, downstream signaling, transcriptional activity, post-translational modification, trafficking and degradation. Primary and immortalized human uterine leiomyoma (HuLM) cells were used for in vitro experiments. Immunodeficient mice xenografted with human leiomyoma tissue explants were used for in vivo studies. Leiomyoma samples were obtained from patients enrolled in an ongoing double-blinded, phase II, randomized controlled trial. Here, we found that simvastatin significantly reduced E2 -induced proliferation and PCNA expression. In addition, simvastatin reduced total ER-α expression in leiomyoma cells and altered its subcellular localization by inhibiting its trafficking to the plasma membrane and nucleus. Simvastatin also inhibited E2 downstream signaling, including ERK and AKT pathways, E2 /ER transcriptional activity and E2 -responsive genes. To explain simvastatin effects on ER-α level and trafficking, we examined its effects on ER-α post-translational processing. We noticed that simvastatin reduced ER-α palmitoylation; a required modification for its stability, trafficking to plasma membrane, and signaling. We also observed an increase in ubiquitin-mediated ER-α degradation. Importantly, we found that the effects of simvastatin on ER-α expression were recapitulated in the xenograft leiomyoma mouse model and human tissues. Thus, our data suggest that simvastatin modulates several E2 /ER signaling targets with potential implications in leiomyoma therapy and beyond., (Copyright © 2021 Elsevier Ltd. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
4. Simvastatin ameliorates altered mechanotransduction in uterine leiomyoma cells.
- Author
-
Afrin S, Islam MS, Patzkowsky K, Malik M, Catherino WH, Segars JH, and Borahay MA
- Subjects
- A Kinase Anchor Proteins drug effects, A Kinase Anchor Proteins genetics, A Kinase Anchor Proteins metabolism, Collagen Type I drug effects, Collagen Type I genetics, Collagen Type I metabolism, Cyclin D1 drug effects, Cyclin D1 genetics, Cyclin D1 metabolism, Extracellular Matrix drug effects, Extracellular Matrix genetics, Extracellular Matrix metabolism, Female, Focal Adhesion Protein-Tyrosine Kinases drug effects, Focal Adhesion Protein-Tyrosine Kinases genetics, Focal Adhesion Protein-Tyrosine Kinases metabolism, Humans, Integrin beta1 drug effects, Integrin beta1 genetics, Integrin beta1 metabolism, Leiomyoma metabolism, Mechanotransduction, Cellular genetics, Minor Histocompatibility Antigens drug effects, Minor Histocompatibility Antigens genetics, Minor Histocompatibility Antigens metabolism, Myosin-Light-Chain Kinase drug effects, Myosin-Light-Chain Kinase genetics, Myosin-Light-Chain Kinase metabolism, Phosphorylation, Primary Cell Culture, Proto-Oncogene Proteins drug effects, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, RNA, Messenger drug effects, RNA, Messenger metabolism, Uterine Neoplasms metabolism, rho-Associated Kinases drug effects, rho-Associated Kinases genetics, rho-Associated Kinases metabolism, rhoA GTP-Binding Protein drug effects, rhoA GTP-Binding Protein genetics, rhoA GTP-Binding Protein metabolism, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Leiomyoma genetics, Mechanotransduction, Cellular drug effects, Simvastatin pharmacology, Uterine Neoplasms genetics
- Abstract
Background: Uterine leiomyomas, the most common tumors of the female reproductive system, are characterized by excessive deposition of disordered stiff extracellular matrix and fundamental alteration in the mechanical signaling pathways. Specifically, these alterations affect the normal dynamic state of responsiveness to mechanical cues in the extracellular environment. These mechanical cues are converted through integrins, cell membrane receptors, to biochemical signals including cytoskeletal signaling pathways to maintain mechanical homeostasis. Leiomyoma cells overexpress β1 integrin and other downstream mechanical signaling proteins. We previously reported that simvastatin, an antihyperlipidemic drug, has antileiomyoma effects through cellular, animal model, and epidemiologic studies., Objective: This study aimed to examine the hypothesis that simvastatin might influence altered mechanotransduction in leiomyoma cells., Study Design: This is a laboratory-based experimental study. Primary leiomyoma cells were isolated from 5 patients who underwent hysterectomy at the Department of Gynecology and Obstetrics of the Johns Hopkins University Hospital. Primary and immortalized human uterine leiomyoma cells were treated with simvastatin at increasing concentrations (0.001, 0.01, 0.1, and 1 μM, or control) for 48 hours. Protein and mRNA levels of β1 integrin and extracellular matrix components involved in mechanical signaling were quantified by quantitative real-time polymerase chain reaction, western blotting, and immunofluorescence. In addition, we examined the effect of simvastatin on the activity of Ras homolog family member A using pull-down assay and gel contraction., Results: We found that simvastatin significantly reduced the protein expression of β1 integrin by 44% and type I collagen by 60% compared with untreated leiomyoma cells. Simvastatin-treated cells reduced phosphorylation of focal adhesion kinase down to 26%-60% of control, whereas it increased total focal adhesion kinase protein expression. Using a Ras homolog family member A pull-down activation assay, we observed reduced levels of active Ras homolog family member A in simvastatin-treated cells by 45%-85% compared with control. Consistent with impaired Ras homolog family member A activation, simvastatin treatment reduced tumor gel contraction where gel area was 122%-153% larger than control. Furthermore, simvastatin treatment led to reduced levels of mechanical signaling proteins involved in β1 integrin downstream signaling, such as A-kinase anchor protein 13, Rho-associated protein kinase 1, myosin light-chain kinase, and cyclin D1., Conclusion: The results of this study suggest a possible therapeutic role of simvastatin in restoring the altered state of mechanotransduction signaling in leiomyoma. Collectively, these findings are aligned with previous epidemiologic studies and other reports and support the need for clinical trials., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
5. Novel effects of simvastatin on uterine fibroid tumors: in vitro and patient-derived xenograft mouse model study.
- Author
-
Borahay MA, Vincent K, Motamedi M, Sbrana E, Kilic GS, Al-Hendy A, and Boehning D
- Subjects
- Animals, Cell Line, Tumor, Estrogens pharmacology, Female, Humans, Immunohistochemistry, In Vitro Techniques, Ki-67 Antigen drug effects, Ki-67 Antigen metabolism, Mice, Phosphorylation drug effects, Progesterone pharmacology, Progestins pharmacology, Proto-Oncogene Proteins c-akt metabolism, Rats, Signal Transduction drug effects, Xenograft Model Antitumor Assays, Apoptosis drug effects, Cell Proliferation drug effects, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Leiomyoma metabolism, Proto-Oncogene Proteins c-akt drug effects, Simvastatin pharmacology, Uterine Neoplasms metabolism
- Abstract
Objective: Uterine leiomyomas represent a common gynecologic problem with no satisfactory long-term medical treatment. The purpose of this study is to examine the effects of simvastatin on uterine leiomyoma, both in vitro and in vivo., Study Design: This is a laboratory-based experimental study. For in vitro studies, we used human and rat leiomyoma cells. For in vivo studies, we used immunodeficient mice supplemented with estrogen/progesterone pellets xenografted with human leiomyoma tissue explant., Results: For in vitro studies, cells were treated with different concentrations of simvastatin for 48 hours. Simvastatin induced dose-dependent apoptosis in leiomyoma cells as measured by a fluorometric caspase-3 activity assay, and inhibited proliferation as demonstrated by an (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) assay (both were significant at 5 and 10 μM). In addition, simvastatin decreased Akt signaling pathway phosphorylation as examined using Western blot analysis. For in vivo studies, animals were treated for 28 days with simvastatin (20 μg/gm body weight/day) vs vehicle control. The treatment inhibited tumor growth as measured weekly using calipers and/ or ultrasound (P < .01). Finally, simvastatin decreased expression of the proliferation marker Ki67 in xenograft tumor tissue as examined by immunohistochemistry (P = .02)., Conclusion: Simvastatin can be a promising treatment for uterine leiomyoma. Further studies, including pharmacokinetic and drug delivery studies, are required., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
6. Simvastatin potently induces calcium-dependent apoptosis of human leiomyoma cells.
- Author
-
Borahay MA, Kilic GS, Yallampalli C, Snyder RR, Hankins GD, Al-Hendy A, and Boehning D
- Subjects
- Blotting, Western, Calcium Channels, L-Type metabolism, Cell Cycle drug effects, Cell Line, Tumor, Cell Survival drug effects, Chelating Agents pharmacology, Cytosol drug effects, Cytosol metabolism, Dose-Response Relationship, Drug, Egtazic Acid analogs & derivatives, Egtazic Acid pharmacology, Humans, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Leiomyoma metabolism, Leiomyoma pathology, MAP Kinase Signaling System drug effects, Microscopy, Fluorescence, Mitogen-Activated Protein Kinases metabolism, Phosphorylation drug effects, Apoptosis drug effects, Calcium metabolism, Cell Proliferation drug effects, Simvastatin pharmacology
- Abstract
Statins are drugs commonly used for the treatment of high plasma cholesterol levels. Beyond these well known lipid-lowering properties, they possess broad-reaching effects in vivo, including antitumor effects. Statins inhibit the growth of multiple tumors. However, the mechanisms remain incompletely understood. Here we show that simvastatin inhibits the proliferation of human leiomyoma cells. This was associated with decreased mitogen-activated protein kinase signaling and multiple changes in cell cycle progression. Simvastatin potently stimulated leiomyoma cell apoptosis in a manner mechanistically dependent upon apoptotic calcium release from voltage-gated calcium channels. Therefore, simvastatin possesses antitumor effects that are dependent upon the apoptotic calcium release machinery., (© 2014 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
7. Simvastatin-loaded liposome nanoparticles treatment for uterine leiomyoma in a patient-derived xenograft mouse model: a pilot study.
- Author
-
El Sabeh, Malak, Vincent, Kathleen L., Afrin, Sadia, Motamedi, Massoud, Saada, Jamal, Yang, Jinping, Ozpolat, Bulent, Kilic, Gokhan S., and Borahay, Mostafa A.
- Abstract
Uterine leiomyomas are complex tumours with limited medical treatment options. Simvastatin is used to treat hypercholesterolaemia and has shown promising effects as a treatment option for leiomyomas. Previously, our group demonstrated a promising effect of simvastatin treatment in a patient-derived xenograft mouse model. Here, we tested the efficacy of simvastatin liposomal nanoparticles (NPs). After bilateral leiomyoma xenograft implantation, mice (N = 12) were divided into three treatment arms: control, simvastatin and simvastatin-loaded liposome NPs (simvastatin-NPs). Treatment with simvastatin significantly reduced tumour volume and inhibited the Ki67 expression when compared to the control group. There was a trend of reduced tumour volume and Ki67 expression after treatment with simvastatin-NP; however, the results were not significant. Due to low bioavailability and short half-life of simvastatin, liposomal NPs have the potential to enhance drug delivery, however, in this study NP did not provide improvement over simvastatin, but did demonstrate their potential for the delivery of simvastatin.Impact statementWhat is already known on this subject? Simvastatin treatment in a patient-derived xenograft mouse model reduced tumour growth and decreased proliferation.What do the results of this study add? Treatment with simvastatin significantly reduced tumour volume and inhibited the Ki67 expression when compared to the control group. There was a trend of reduced tumour volume and Ki67 expression after treatment with simvastatin-NP, however, it did not improve the efficacy of simvastatin at reducing tumour growth and proliferation.What are the implications of these findings for clinical practice and/or further research? More studies are needed to optimise the formulation of NPs to further enhance the sustainable delivery of simvastatin. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
8. Simvastatin inhibits stem cell proliferation in human leiomyoma via TGF‐β3 and Wnt/β‐Catenin pathways.
- Author
-
Afrin, Sadia, Ali, Mohamed, El Sabeh, Malak, Yang, Qiwei, Al‐Hendy, Ayman, and Borahay, Mostafa A.
- Subjects
HUMAN stem cells ,INHIBITION of cellular proliferation ,STEM cell niches ,SIMVASTATIN ,UTERINE fibroids ,WNT signal transduction - Abstract
Uterine leiomyoma (UL) is the most common gynaecologic tumour, affecting an estimated 70 to 80% of women. Leiomyomas develop from the transformation of myometrial stem cells into leiomyoma stem (or tumour‐initiating) cells. These cells undergo self‐renewal and differentiation to mature cells, both are necessary for the maintenance of tumour stem cell niche and tumour growth, respectively. Wnt/β‐catenin and TGF‐β/SMAD pathways, both overactive in UL, promote stem cell self‐renewal, crosstalk between stem and mature cells, cellular proliferation, extracellular matrix (ECM) accumulation and drive overall UL growth. Recent evidence suggests that simvastatin, an antihyperlipidemic drug, may have anti‐leiomyoma properties. Herein, we investigated the effects of simvastatin on UL stem cells. We isolated leiomyoma stem cells by flow cytometry using DyeCycle Violet staining and Stro‐1/CD44 surface markers. We found that simvastatin inhibits proliferation and induces apoptosis in UL stem cells. In addition, it also suppressed the expression of the stemness markers Nanog, Oct4 and Sox2. Simvastatin significantly decreased the production of the key ECM proteins, collagen 1 and fibronectin. Finally, it inhibited genes and/or proteins expression of TGF‐β1, 2 and 3, SMAD2, SMAD4, Wnt4, β‐Catenin, LRP6, AXIN2 and Cyclin D1 in UL stem cells, all are key drivers of the TGF‐β3/SMAD2 and Wnt4/β‐Catenin pathways. Thus, we have identified a novel stem cell‐targeting anti‐leiomyoma simvastatin effect. Further studies are needed to replicate these findings in vivo. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
9. Simvastatin Inhibits Wnt/β-Catenin Pathway in Uterine Leiomyoma.
- Author
-
Sabeh, Malak El, Saha, Subbroto Kumar, Afrin, Sadia, and Borahay, Mostafa A
- Subjects
SIMVASTATIN ,UTERINE fibroids ,PROTEIN expression - Abstract
The Wnt/β-catenin pathway is upregulated in uterine leiomyomas, the most common benign tumors in the female reproductive tract. Simvastatin is an antihyperlipidemic drug, and previous in vitro and in vivo reports showed that it may have therapeutic effects in treating leiomyomas. The objective of this study was to examine the effects of simvastatin on the Wnt/β-catenin signaling pathway in leiomyoma. We treated primary and immortalized human leiomyoma cells with simvastatin and examined its effects using quantitative real-time polymerase chain reaction, Western blotting, and immunocytochemistry. We also examined the effects using human leiomyoma tissues from an ongoing randomized controlled trial in which women with symptomatic leiomyoma received simvastatin (40 mg) or placebo for 3 months prior to their surgery. The results of this study revealed that simvastatin significantly reduced the expression of Wnt4 and its co-receptor LRP5. After simvastatin treatment, levels of total β-catenin and its active form, nonphosphorylated β-catenin, were reduced in both cell types. Additionally, simvastatin reduced the expression of Wnt4 and total β-catenin, as well as nonphosphorylated β-catenin protein expression in response to estrogen and progesterone. Simvastatin also inhibited the expression of c-Myc, a downstream target of the Wnt/β-catenin pathway. The effect of simvastatin on nonphosphorylated-β-catenin, the key regulator of the Wnt/β-catenin pathway, was recapitulated in human leiomyoma tissue. These results suggest that simvastatin may have a beneficial effect on uterine leiomyoma through suppressing the overactive Wnt/β-catenin pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
10. Simvastatin, at clinically relevant concentrations, affects human uterine leiomyoma growth and extracellular matrix production.
- Author
-
Malik, Minnie, Britten, Joy, Borahay, Mostafa, Segars, James, and Catherino, William H
- Subjects
- *
APOPTOSIS , *CELL physiology , *CELLULAR signal transduction , *COLLAGEN , *COMPARATIVE studies , *DOSE-effect relationship in pharmacology , *EXTRACELLULAR space , *GLYCOPROTEINS , *RESEARCH methodology , *MEDICAL cooperation , *MYOMETRIUM , *RESEARCH , *UTERINE fibroids , *UTERINE tumors , *EVALUATION research , *SIMVASTATIN , *CANCER cell culture , *PHARMACODYNAMICS , *METABOLISM - Abstract
Objective: To observe the antifibroid effects of therapeutic concentrations of simvastatin, which interferes with cholesterol biosynthesis, a known precursor of five major classes of steroid hormones, including progesterone and estrogen, which play a major role in the development and growth of uterine leiomyomas.Design: Two-dimensional and three-dimensional cell culture study of immortalized human leiomyoma and patient-matched myometrium cells treated with simvastatin.Setting: University laboratory.Patient(s): None.Interventions(s): None.Main Outcome Measure(s): Cell proliferation, alteration in apoptotic signaling pathways, and extracellular matrix (ECM) protein production.Result(s): Simvastatin demonstrated a concentration-dependent antiproliferative effect on both the leiomyoma cells and the patient-matched myometrium cells, but a higher inhibitory effect at lower concentrations of simvastatin was observed in leiomyoma cells. Simvastatin also regulated leiomyoma cell apoptosis through a concentration-dependent increase in activity of caspase-3. Simvastatin significantly inhibited expression of major ECM proteins collagen I, collagen III, fibronectin, versican, and brevican in leiomyoma cells at concentrations as low as 10-9 mol/L within 48 hours of exposure.Conclusion(s): Simvastatin induces apoptosis in uterine leiomyoma cells at low concentrations, as evidenced by increased active caspase levels. Furthermore, inhibited production of the ECM proteins may lead to reduction in tumor size. Simvastatin may represent a novel therapeutic treatment strategy for uterine leiomyomas. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
11. Simvastatin reduces plasma membrane caveolae and caveolin-1 in uterine leiomyomas.
- Author
-
Afrin, Sadia, El Sabeh, Malak, Miyashita-Ishiwata, Mariko, Charewycz, Natasha, Singh, Bhuchitra, and Borahay, Mostafa A.
- Subjects
- *
UTERINE fibroids , *CELL membranes , *SIMVASTATIN , *PROTEIN kinase B , *CAVEOLAE , *ESTROGEN receptors - Abstract
Uterine leiomyomas, or fibroids, are estrogen dependent benign tumor in women, however, they have limited treatment options. Simvastatin, a drug commonly used to treat high cholesterol. Recently we demonstrated that simvastatin alters estrogen signaling by reducing the expression and trafficking of the estrogen receptor-α (ER-α) in human uterine leiomyoma cells. Caveolae are invaginations of the plasma membrane where ER-α is known to localize and directly interacts with the caveolar protein caveolin-1 (CAV1). This study examines the effects of simvastatin on plasma membrane caveolae and the expression and palmitoylation of CAV1 in human leiomyomas which may influence ER-α signaling. We performed in vitro experiments using primary and immortalized human uterine leiomyoma cells. The caveolae were quantified using transmission electron microscopy. Additionally, we examined the impact of simvastatin treatment (40 mg orally per day for 12 weeks) on human leiomyoma tissue obtained from a randomized controlled trial. The CAV1 protein and mRNA levels were determined using quantitative real-time polymerase chain reactions, western blotting, and immunofluorescence analyses. Simvastatin decreased the number of caveolae in primary leiomyoma cells and reduced CAV1 abundance in whole cells and remarkably the plasma protein fraction. It also decreased CAV1 palmitoylation, a post-translational modification associated with CAV1 activation. The effects of simvastatin on CAV1 were recapitulated in human leiomyoma tissue samples. Our results identify caveolae and CAV1 as novel targets of simvastatin which may contribute to the recently described effects of simvastatin on ER-α signaling and plasma membrane trafficking. Simvastatin decreases caveolae formation and CAV1 palmitoylation in leiomyoma cells and reduces CAV1 abundance in both leiomyoma cells and human tissue, thus highlighting these membrane components as new targets of simvastatin. Palm, palmitate; CAV1, caveolin-1; ER-α, estrogen receptor α, PI3K, phosphoinositide 3-kinase; AKT, protein kinase B; MAPK, mitogen-activated protein kinase. [Display omitted] [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
12. SIMVASTATIN SUPPRESSES PROLIFERATION, EXTRACELLULAR MATRIX ACCUMULATION AND Wnt4/Β-CATENIN PATHWAYS IN HUMAN LEIOMYOMA STEM CELLS.
- Author
-
Afrin, Sadia, Ali, Mohamed, El Sabeh, Malak, Yang, Qiwei, Al-Hendy, Ayman, and Borahay, Mostafa A.
- Subjects
- *
HUMAN stem cells , *EXTRACELLULAR matrix , *SIMVASTATIN - Published
- 2021
- Full Text
- View/download PDF
13. Effect of simvastatin on integrin-β1 and its downstream mediators in human leiomyoma cells.
- Author
-
Afrin, Sadia, Islam, Soriful, Su, Szu-Chi, and Borahay, Mostafa A.
- Subjects
- *
SIMVASTATIN , *UTERINE fibroids , *HUMAN beings - Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.