Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Biol Reprod ; 108(2): 172-182, 2023 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-36173920

RESUMO

Coronavirus disease 2019 (COVID-19) is a multi-system disease that has led to a pandemic with unprecedented ramifications. The pandemic has challenged scientists for the past 2 years and brought back previously abandoned research topics. COVID-19 infection causes a myriad of symptoms ranging from mild flu-like symptoms to severe illness requiring hospitalization. Case reports showed multiple systemic effects of COVID-19 infection, including acute respiratory distress syndrome, fibrosis, colitis, thyroiditis, demyelinating syndromes, and mania, indicating that COVID-19 can affect most human body systems. Unsurprisingly, a major concern for women all over the globe is whether a COVID-19 infection has any long-term effects on their menstrual cycle, fertility, or pregnancy. Published data have suggested an effect on the reproductive health, and we hypothesize that the reported reproductive adverse effects are due to the robust immune reaction against COVID-19 and the associated cytokine storm. While the COVID-19 receptor (angiotensin converting enzyme, ACE2) is expressed in the ovaries, uterus, vagina, and placenta, we hypothesize that it plays a less important role in the adverse effects on the reproductive system. Cytokines and glucocorticoids act on the hypothalamo-pituitary gonadal axis, arachidonic acid pathways, and the uterus, which leads to menstrual disturbances and pregnancy-related adverse events such as preterm labor and miscarriages. This hypothesis is further supported by the apparent lack of long-term effects on the reproductive health in females, indicating that when the cytokine storm and its effects are dampened, the reproductive health of women is no longer affected.


Assuntos
COVID-19 , Genitália Feminina , Feminino , Humanos , Recém-Nascido , Gravidez , COVID-19/complicações , COVID-19/imunologia , Síndrome da Liberação de Citocina/metabolismo , Genitália Feminina/patologia , Imunidade , SARS-CoV-2
2.
J Cell Mol Med ; 26(5): 1684-1698, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35118811

RESUMO

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/ß-catenin and TGF-ß/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-ß1, 2 and 3, SMAD2, SMAD4, Wnt4, ß-Catenin, LRP6, AXIN2 and Cyclin D1 in UL stem cells, all are key drivers of the TGF-ß3/SMAD2 and Wnt4/ß-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.


Assuntos
Leiomioma , Neoplasias Uterinas , Proliferação de Células/genética , Feminino , Humanos , Leiomioma/tratamento farmacológico , Leiomioma/genética , Sinvastatina/farmacologia , Fator de Crescimento Transformador beta3/metabolismo , Neoplasias Uterinas/genética , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patologia , beta Catenina/metabolismo
3.
J Obstet Gynaecol ; 42(6): 2139-2143, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35166183

RESUMO

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.


Assuntos
Leiomioma , Nanopartículas , Animais , Modelos Animais de Doenças , Xenoenxertos , Humanos , Antígeno Ki-67 , Leiomioma/tratamento farmacológico , Leiomioma/patologia , Lipossomos , Camundongos , Projetos Piloto , Sinvastatina/farmacologia
4.
Reproduction ; 162(2): R1-R18, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34034234

RESUMO

Fibroids are benign tumors caused by the proliferation of myometrial smooth muscle cells in the uterus that can lead to symptoms such as abdominal pain, constipation, urinary retention, and infertility. While traditionally thought of as a disease process intrinsic to the uterus, accumulating evidence suggests that fibroid growth may be linked with the systemic vasculature system, although cell-intrinsic factors are certainly of principal importance in their inception. Fibroids are associated with essential hypertension and preeclampsia, as well as atherosclerosis, for reasons that are becoming increasingly elucidated. Factors such as the renin-angiotensin-aldosterone system, estrogen, and endothelial dysfunction all likely play a role in fibroid pathogenesis. In this review, we lay out a framework for reconceptualizing fibroids as a systemic vascular disorder, and discuss how pharmaceutical agents and other interventions targeting the vasculature may aid in the novel treatment of fibroids.


Assuntos
Leiomioma/fisiopatologia , Doenças Vasculares/patologia , Animais , Feminino , Humanos , Gravidez , Doenças Vasculares/etiologia
5.
Mol Cell Biochem ; 476(9): 3513-3536, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33999334

RESUMO

Uterine leiomyoma is the most common tumor of the female reproductive system and originates from a single transformed myometrial smooth muscle cell. Despite the immense medical, psychosocial, and financial impact, the exact underlying mechanisms of leiomyoma pathobiology are poorly understood. Alterations of signaling pathways are thought to be instrumental in leiomyoma biology. Wnt/ß-catenin pathway appears to be involved in several aspects of the genesis of leiomyomas. For example, Wnt5b is overexpressed in leiomyoma, and the Wnt/ß-catenin pathway appears to mediate the role of MED12 mutations, the most common mutations in leiomyoma, in tumorigenesis. Moreover, Wnt/ß-catenin pathway plays a paracrine role where estrogen/progesterone treatment of mature myometrial or leiomyoma cells leads to increased expression of Wnt11 and Wnt16, which induces proliferation of leiomyoma stem cells and tumor growth. Constitutive activation of ß-catenin leads to myometrial hyperplasia and leiomyoma-like lesions in animal models. Wnt/ß-catenin signaling is also closely involved in mechanotransduction and extracellular matrix regulation and relevant alterations in leiomyoma, and crosstalk is noted between Wnt/ß-catenin signaling and other pathways known to regulate leiomyoma development and growth such as estrogen, progesterone, TGFß, PI3K/Akt/mTOR, Ras/Raf/MEK/ERK, IGF, Hippo, and Notch signaling. Finally, evidence suggests that inhibition of the canonical Wnt pathway using ß-catenin inhibitors inhibits leiomyoma cell proliferation. Understanding the molecular mechanisms of leiomyoma development is essential for effective treatment. The specific Wnt/ß-catenin pathway molecules discussed in this review constitute compelling candidates for therapeutic targeting.


Assuntos
Antineoplásicos/uso terapêutico , Leiomioma/tratamento farmacológico , Terapia de Alvo Molecular , Neoplasias Uterinas/tratamento farmacológico , Via de Sinalização Wnt/efeitos dos fármacos , Animais , Feminino , Humanos , Leiomioma/metabolismo , Leiomioma/patologia , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patologia
6.
Pharmacol Res ; 172: 105856, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34461224

RESUMO

Uterine leiomyomas or fibroids are the most common tumors of the female reproductive tract. Estrogen (E2), 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.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Leiomioma/metabolismo , Sinvastatina/farmacologia , Neoplasias Uterinas/metabolismo , Adolescente , Adulto , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Método Duplo-Cego , Feminino , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Leiomioma/tratamento farmacológico , Lipoilação , Camundongos , Pessoa de Meia-Idade , Transporte Proteico , Proteólise , Transdução de Sinais/efeitos dos fármacos , Sinvastatina/uso terapêutico , Neoplasias Uterinas/tratamento farmacológico , Adulto Jovem
7.
J Biosoc Sci ; 53(3): 356-366, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-32398189

RESUMO

Consanguineous marriage is a deeply rooted tradition in the Arab world. Such marriages are linked to higher rates of recessive genetic diseases. During the Syrian conflict, which started in 2011, around one million Syrian individuals became refugees in Lebanon. This study assessed the consanguinity rates among Syrian refugees living in Lebanon up to three successive consanguineous generations, and examined refugees' awareness of the possible consequences of consanguineous marriage and their attitudes towards consanguinity. Their knowledge of, and access to, premarital screening was also assessed. The study was conducted between January and May 2018. Several study sites representing refugees' distribution within the country were chosen. The study sample included 1008 interviewees from different families. Of those interviewed, 51.9% were in a consanguineous marriage. Interestingly, 23.9% were the product of consanguineous marriages themselves, and 17.9% were consanguineous for three successive generations. The interviewees generally knew about premarital screening, but the majority (61.9%) had not had the screening. The high rates of consanguinity in these Syrian refugees call for immediate action, including raising genetic awareness and providing appropriate genetic counselling. Despite the respondents' familiarity with premarital screening, there was a low rate of uptake of the test, underscoring the importance of providing better education to these refugees.


Assuntos
Refugiados , Consanguinidade , Escolaridade , Humanos , Líbano , Síria
9.
Vaccine X ; 19: 100501, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38832342

RESUMO

Background: In initial COVID-19 clinical trials, menstrual health was not formally monitored, yet anecdotal reports of menstruation changes surfaced on social media. This study aims to assess the association between COVID-19 vaccines and menstruation using Clue, a period-tracking application. Study design: A survey assessing demographics, menstrual health, stress levels, and COVID-19 vaccination was sent to Clue users between 12/7/2021 and 2/9/2022. Inclusion criteria were (1) 18 years or older (2) currently menstruating (3) not pregnant or breastfeeding since 1/2020. Menstrual data was collected for each participant. Users with cycle lengths more than 90 days were excluded. Cycle lengths were calculated for the 6-month average pre-vaccination (PRIOR), the cycle during which vaccination was administered (DURING), the cycle following DURING (AFTER1), and the cycle following AFTER1 (AFTER2). For periods, individuals were stratified based on whether vaccination was received during their menstrual period (DURING). Period lengths were additionally calculated for the 6-month average pre-vaccination (PRIOR), the first period following vaccination (AFTER1), and the period following AFTER1 (AFTER2). For unvaccinated participants, an index date (4/1/2022) was used to similarly designate menstrual cycles and periods. For each participant, cycle length changes for DURING, AFTER1, and AFTER2 compared to PRIOR were determined. Student's t-test compared the mean of these changes between vaccinated and unvaccinated groups. Results: Of 7,559 participants, 6,897 (91 %) were vaccinated. Compared to PRIOR, individuals vaccinated during their menstrual period demonstrated a statistically significant increase in the DURING period length, but not AFTER1 (p = 0.463) and AFTER2 (p = 0.692). No statistically significant changes were observed in period lengths of those vaccinated in between periods or in cycle lengths overall. Conclusion: A small but statistically significant change in period length was observed only in individuals vaccinated for COVID-19 during their menstrual period. Providers can better counsel menstruating individuals to reduce vaccine misinformation.

10.
Life Sci ; 304: 120708, 2022 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-35705139

RESUMO

AIMS: 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. MAIN METHODS: 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. KEY FINDINGS: 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. SIGNIFICANCE: 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.


Assuntos
Cavéolas , Leiomioma , Cavéolas/metabolismo , Caveolina 1/genética , Membrana Celular/metabolismo , Estrogênios/metabolismo , Feminino , Humanos , Leiomioma/tratamento farmacológico , Leiomioma/metabolismo , Sinvastatina/farmacologia
11.
Free Radic Res ; 56(2): 163-172, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35377824

RESUMO

Uterine leiomyomas, the most common tumors of the female reproductive system, are known to have a hypoxic microenvironment. However, the role of such environment in leiomyoma pathobiology remains unknown. The objective was to determine the effects of hypoxia on leiomyoma cells, and the mechanisms. We found that hypoxia induces proliferation and inhibits apoptosis in human leiomyoma cells. This pro-proliferative effect was accompanied by an increase in reactive oxygen species (ROS) generation and the expression of NADPH oxidase 4 (NOX4). The specific NOX4 inhibitor GLX351322 abrogated this hypoxia-induced ROS generation, cellular proliferation, and apoptosis inhibition. To further investigate the mechanism of NOX4-mediated proliferation, we treated leiomyoma cells grown in normoxia with media from leiomyoma cells cultured under hypoxia. This resulted in increased ROS generation and NOX4 expression, suggesting the hypoxia-induced effects are mediated by an autocrine mechanism. We worked to identify the nature of this autocrine factor. We found that the expression of TGF-ß3 and its downstream signaling target pSmad3, are increased in hypoxic leiomyoma cells. To examine the hypothesis that TGF-ß3 is, at least, a part of this autocrine mechanism, we treated hypoxic leiomyoma cells with the HIF-1α inhibitor KC7F2 which we discovered to ameliorate the hypoxia-induced TGF-ß3 expression. Furthermore, pharmacologic inhibition with the TGF-ß/Smad inhibitor SB431542 reduced hypoxia-induced NOX4 expression and ROS generation and attenuated cell proliferation. Thus, we have identified a novel mechanism by which hypoxia induces proliferation in leiomyoma cells. This finding adds to our understanding of leiomyoma pathobiology and can help in identifying new therapeutic targets.


Assuntos
Leiomioma , Fator de Crescimento Transformador beta3 , Proliferação de Células , Feminino , Humanos , Hipóxia , Leiomioma/genética , NADPH Oxidase 4/genética , NADPH Oxidase 4/metabolismo , NADPH Oxidases/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Fator de Crescimento Transformador beta3/metabolismo , Fator de Crescimento Transformador beta3/farmacologia , Microambiente Tumoral
12.
Life Sci ; 290: 120238, 2022 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-34942165

RESUMO

AIMS: Recent evidence suggests that repetitive hypoxia occurs during menstrual cycles due to vasoconstriction and myometrial contraction. It is unknown if hypoxia contributes to the development of uterine leiomyoma, the most common tumor of the female reproductive system. This study aims to characterize the response to hypoxia in leiomyoma and myometrial cells; and determine if an aberrant leiomyoma response to hypoxia may contribute to leiomyomatogenesis. MAIN METHODS: Primary and immortalized leiomyoma and myometrial cells were cultured under normoxic and hypoxic conditions. Expression levels of vascular endothelial growth factor-A (VEGF-A), adrenomedullin (ADM), endothelin-1 (ET-1), and hypoxia-inducible factor-1 alpha (HIF-1α) were measured by qRT-PCR, western blotting and ELISA. Cell proliferation was assessed using MTT assay and proliferating-cell-nuclear-antigen (PCNA) expression. KC7F2 (HIF-1α inhibitor) was used to examine the regulating mechanisms. KEY FINDINGS: As expected, hypoxia induced HIF-1α expression in both leiomyoma and myometrial cells. However, hypoxia induced VEGF-A, ET-1 and ADM expression and VEGF-A secretion into the culture media in leiomyoma but not myometrial cells. MTT assay and PCNA expression showed that hypoxia induces proliferation in leiomyoma, but not myometrial cells. HIF-1α inhibitor abrogated the hypoxia-induced VEGF-A, ET-1, ADM, and PCNA expression in leiomyoma cells. SIGNIFICANCE: This study suggests an aberrant leiomyoma cellular response to hypoxia compared to myometrium. This differential response to menstruation-related repetitive hypoxia episodes may lead to selective proliferation of hypoxia-adaptive leiomyoma cells and contribute to leiomyoma growth. Thus, in addition to adding to our understanding of leiomyoma pathobiology, the study proposes angiogenic factors as a potential leiomyoma therapeutic target.


Assuntos
Hipóxia Celular/fisiologia , Leiomioma/metabolismo , Miométrio/metabolismo , Adrenomedulina/análise , Linhagem Celular , Proliferação de Células , Endotelina-1/análise , Feminino , Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/análise , Leiomioma/patologia , Miométrio/patologia , Antígeno Nuclear de Célula em Proliferação/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/análise
13.
Reprod Sci ; 28(11): 3285-3288, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34008153

RESUMO

Uterine fibroids are the most common tumors of the female reproductive tract, affecting up to 80% of women. Despite their heavy burden and high prevalence, available medical treatment options are limited and are offered to patients assuming equal responsiveness. These benign tumors are complex, originating from potentially diverse pathobiologic processes, yet they are all managed in a rather standardized symptom-oriented approach that does not take into account the underlying processes. With our increasing understanding of the interplay between genes, epigenetics, individual's lifestyle, and the environment in disease development, uterine fibroid management should be geared towards individualized preventive and treatment options. For example, it seems that some subsets of patients with fibroids also suffer from vitamin D deficiency, hypertension, metabolic syndrome, or other conditions. It is possible that these subsets may have different underlying processes and different responsiveness to different treatment options. Herein, we call for a futuristic paradigm shift of research to develop a new model to manage uterine fibroids with the treatment approach varying depending on the patient's perceived underlying processes as assessed by medical, social, family history, and relevant investigations. This is only possible through the collaboration of scientists, physicians, and funding agencies and with the help of our patients.


Assuntos
Leiomioma/diagnóstico , Leiomioma/terapia , Medicina de Precisão/métodos , Neoplasias Uterinas/diagnóstico , Neoplasias Uterinas/terapia , Ensaios Clínicos como Assunto/métodos , Feminino , Humanos , Leiomioma/prevenção & controle , Neoplasias Uterinas/prevenção & controle
14.
Endocrinology ; 162(12)2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34614511

RESUMO

The Wnt/ß-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/ß-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 ß-catenin and its active form, nonphosphorylated ß-catenin, were reduced in both cell types. Additionally, simvastatin reduced the expression of Wnt4 and total ß-catenin, as well as nonphosphorylated ß-catenin protein expression in response to estrogen and progesterone. Simvastatin also inhibited the expression of c-Myc, a downstream target of the Wnt/ß-catenin pathway. The effect of simvastatin on nonphosphorylated-ß-catenin, the key regulator of the Wnt/ß-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/ß-catenin pathway.


Assuntos
Leiomioma/patologia , Sinvastatina/farmacologia , Neoplasias Uterinas/patologia , Via de Sinalização Wnt/efeitos dos fármacos , Adolescente , Adulto , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cultivadas , Ensaios Clínicos Fase II como Assunto , Método Duplo-Cego , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Feminino , Humanos , Leiomioma/genética , Leiomioma/metabolismo , Pessoa de Meia-Idade , Cultura Primária de Células , Ensaios Clínicos Controlados Aleatórios como Assunto , Neoplasias Uterinas/genética , Neoplasias Uterinas/metabolismo , Adulto Jovem , beta Catenina/antagonistas & inibidores , beta Catenina/metabolismo
15.
Stem Cell Rev Rep ; 17(3): 803-820, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33155150

RESUMO

Stem cells in the endometrium and myometrium possess an immense regenerative potential which is necessary to maintain the menstrual cycle and support pregnancy. These cells, as well as bone marrow stem cells, have also been implicated in the development of common benign gynecological disorders including leiomyomas, endometriosis and adenomyosis. Current evidence suggests the conversion of uterine stem cells to tumor initiating stem cells in leiomyomas, endometriosis stem cells, and adenomyosis stem cells, acquiring genetic and epigenetic alterations for the progression of each benign condition. In this comprehensive review, we aim to summarize the progress that has been made to characterize the involvement of stem cells in the pathogenesis of benign gynecologic conditions which, despite their enormous burden, are not yet fully understood. We focus on the stem cell characteristics and aberrations that contribute to the development of benign gynecological disorders and the possible clinical implications of what is known so far. Lastly, we discuss the role of uterine stem cells in the setting of regenerative medicine, particularly in the treatment of Asherman syndrome.Graphical abstract.


Assuntos
Adenomiose , Endometriose , Leiomioma , Adenomiose/patologia , Endometriose/patologia , Endometriose/terapia , Feminino , Humanos , Leiomioma/patologia , Leiomioma/terapia , Miométrio/patologia , Células-Tronco Neoplásicas/patologia , Gravidez
16.
Nutrients ; 13(6)2021 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-34063835

RESUMO

A healthy lifestyle and a balanced diet play a paramount role in promoting and maintaining homeostatic functions and preventing an array of chronic and debilitating diseases. Based upon observational and epidemiological investigations, it is clear that nutritional factors and dietary habits play a significant role in gynecological disease development, including uterine leiomyoma, endometriosis, polycystic ovary syndrome, and gynecological malignancies. Diets rich in fruits and vegetables, Mediterranean diets, green tea, vitamin D, and plant-derived natural compounds may have a long-term positive impact on gynecological diseases, while fats, red meat, alcohol, and coffee may contribute to their development. Data regarding the association between dietary habits and gynecological disorders are, at times, conflicting, with potential confounding factors, including food pollutants, reduced physical activity, ethnic background, and environmental factors limiting overall conclusions. This review provides a synopsis of the current clinical data and biological basis of the association between available dietary and nutritional data, along with their impact on the biology and pathophysiology of different gynecological disorders, as well as an outlook on future directions that will guide further investigational research.


Assuntos
Dieta/efeitos adversos , Comportamento Alimentar/fisiologia , Doenças dos Genitais Femininos/etiologia , Doenças dos Genitais Femininos/fisiopatologia , Estado Nutricional/fisiologia , Feminino , Humanos
17.
J Clin Endocrinol Metab ; 106(2): e990-e1001, 2021 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-33280041

RESUMO

CONTEXT: Questions regarding the superiority of free and bioavailable 25-hydroxyvitamin D [25(OH)D] in predicting health outcomes remain unresolved. OBJECTIVE: This study investigates the impact of vitamin D variables-total, bioavailable, or free 25(OH)D-on indices of bone and mineral metabolism, at baseline and in response to 2 vitamin D doses. DESIGN: Our objectives are implemented as exploratory analyses on data collected in a 1-year, double-blind, randomized controlled trial completed in July 2014. SETTING: Participants were recruited from 3 major hospitals in an ambulatory setting. PARTICIPANTS: Participants were >65 years of age, overweight, and had a baseline serum 25(OH)D between 10 and 30 ng/mL. A total of 221 participants completed the study. INTERVENTION: Subjects were randomized to receive calcium and oral vitamin D3 (600 IU/day or 3750 IU/day) supplementation. RESULTS: Participants who received the higher vitamin D dose had levels that were 1.3- to 1.4-fold higher than those taking the lower dose, for all variables (P value < 0.001). Serum values of bioavailable and free 25(OH)D were associated with total 25(OH)D, with r values of 0.942 and 0.943, respectively (P value < 0.001). Parathyroid hormone (PTH) was negatively associated with all vitamin D variables, with correlation coefficients ranging from -0.22 to -0.25, while calcium and bone turnover markers (carboxy-terminal collagen crosslinks and osteocalcin) did not. Only total 25(OH)D had a positive relationship with % change bone mineral density (BMD) at the femoral neck at 12 months, while only free and bioavailable 25(OH) had a positive relationship with % change total body BMD at 12 months. CONCLUSION: Calculated free and bioavailable 25(OH)D do not appear to be superior to total 25(OH)D in predicting indices of bone health in an elderly population.


Assuntos
Biomarcadores/sangue , Densidade Óssea , Vitamina D/análogos & derivados , Vitaminas/sangue , Idoso , Método Duplo-Cego , Feminino , Seguimentos , Humanos , Masculino , Prognóstico , Vitamina D/sangue
18.
Placenta ; 90: 90-97, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-32056558

RESUMO

INTRODUCTION: The placenta is a transitory organ essential for proper fetal maturation and growth. Trophoblasts, the main cell type of the placenta, differentiate along the villous or extravillous pathways. The ability of villous cytotrophoblasts to undergo an epithelial-to-mesenchymal transition to form the invasive extravillous trophoblasts is vital for a successful pregnancy outcome. Many trophoblastic cell lines, including HTR-8/SVneo, have been widely used to investigate extravillous trophoblast biology and functions. We have previously reported that HTR-8/SVneo cell line contains a mixed populations of epithelial and mesenchymal cells. Uncovering the mechanisms underlying this heterogeneity is essential for the proper study of normal and pathological placental function. METHODS: HTR-8/SVneo was subjected to monoclonal isolation, spheroid formation assay and cell sorting to isolate pure epithelial and mesenchymal populations. These fractions were maintained in culture and assessed for expression of epithelial and mesenchymal markers using quantitative real-time PCR and immunofluorescence. In addition, the implication of TGFß in the EMT process was investigated using a selective inhibitor of TGF-ßR1 (A83-01). RESULTS: Passaging of the pure epithelial population maintained under normal culture condition resulted in a shift to a mesenchymal phenotype. This transition was reduced upon inhibiting TGF-ßR1. Similarly, E-cadherin positive HTR-8/SVneo spheroids plated in 2D culture resulted in the emergence of streams of invading mesenchymal cells. DISCUSSION: HTR-8/SVneo cell line is undergoing EMT under normal culture condition and TGFß is a key mediator of this process. Our results raise the possibility of using HTR-8/SVneo cell line as a model to investigate EMT in extravillous trophoblast cells.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Placenta/citologia , Trofoblastos/citologia , Caderinas/metabolismo , Linhagem Celular , Feminino , Humanos , Placenta/metabolismo , Gravidez , Trofoblastos/metabolismo
20.
F S Sci ; 2(2): 114-115, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34355207
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA