Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Hum Reprod ; 39(5): 1023-1041, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38511208

RESUMO

STUDY QUESTION: How does ovarian stimulation (OS), which is used to mature multiple oocytes for ART procedures, impact the principal cellular compartments and transcriptome of the human endometrium in the periovulatory and mid-secretory phases? SUMMARY ANSWER: During the mid-secretory window of implantation, OS alters the abundance of endometrial immune cells, whereas during the periovulatory period, OS substantially changes the endometrial transcriptome and impacts both endometrial glandular and immune cells. WHAT IS KNOWN ALREADY: Pregnancies conceived in an OS cycle are at risk of complications reflective of abnormal placentation and placental function. OS can alter endometrial gene expression and immune cell populations. How OS impacts the glandular, stromal, immune, and vascular compartments of the endometrium, in the periovulatory period as compared to the window of implantation, is unknown. STUDY DESIGN, SIZE, DURATION: This prospective cohort study carried out between 2020 and 2022 included 25 subjects undergoing OS and 25 subjects in natural menstrual cycles. Endometrial biopsies were performed in the proliferative, periovulatory, and mid-secretory phases. PARTICIPANTS/MATERIALS, SETTING, METHODS: Blood samples were processed to determine serum estradiol and progesterone levels. Both the endometrial transcriptome and the principal cellular compartments of the endometrium, including glands, stroma, immune, and vasculature, were evaluated by examining endometrial dating, differential gene expression, protein expression, cell populations, and the three-dimensional structure in endometrial tissue. Mann-Whitney U tests, unpaired t-tests or one-way ANOVA and pairwise multiple comparison tests were used to statistically evaluate differences. MAIN RESULTS AND THE ROLE OF CHANCE: In the periovulatory period, OS induced high levels of differential gene expression, glandular-stromal dyssynchrony, and an increase in both glandular epithelial volume and the frequency of endometrial monocytes/macrophages. In the window of implantation during the mid-secretory phase, OS induced changes in endometrial immune cells, with a greater frequency of B cells and a lower frequency of CD4 effector T cells. LARGE SCALE DATA: The data underlying this article have been uploaded to the Genome Expression Omnibus/National Center for Biotechnology Information with accession number GSE220044. LIMITATIONS, REASONS FOR CAUTION: A limited number of subjects were included in this study, although the subjects within each group, natural cycle or OS, were homogenous in their clinical characteristics. The number of subjects utilized was sufficient to identify significant differences; however, with a larger number of subjects and additional power, we may detect additional differences. Another limitation of the study is that proliferative phase biopsies were collected in natural cycles, but not in OS cycles. Given that the OS cycle subjects did not have known endometrial factor infertility, and the comparisons involved subjects who had a similar and robust response to stimulation, the findings are generalizable to women with a normal response to OS. WIDER IMPLICATIONS OF THE FINDINGS: OS substantially altered the periovulatory phase endometrium, with fewer transcriptomic and cell type-specific changes in the mid-secretory phase. Our findings show that after OS, the endometrial microenvironment in the window of implantation possesses many more similarities to that of a natural cycle than does the periovulatory endometrium. Further investigation of the immune compartment and the functional significance of this cellular compartment under OS conditions is warranted. STUDY FUNDING/COMPETING INTERESTS: Research reported in this publication was supported by the National Institute of Allergy and Infectious Diseases (R01AI148695 to A.M.B. and N.C.D.), Eunice Kennedy Shriver National Institute of Child Health and Human Development (R01HD109152 to R.A.), and the March of Dimes (5-FY20-209 to R.A.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health or March of Dimes. All authors declare no conflict of interest.


Assuntos
Endométrio , Indução da Ovulação , Transcriptoma , Humanos , Feminino , Endométrio/metabolismo , Adulto , Microambiente Celular , Estudos Prospectivos , Estradiol/sangue , Implantação do Embrião/fisiologia , Progesterona/sangue , Progesterona/metabolismo , Gravidez , Ciclo Menstrual
2.
Reproduction ; 166(2): 161-174, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37252830

RESUMO

In brief: Endometrial stromal cell motility is fundamental to regeneration and repair of this tissue and crucial for successful reproduction. This paper shows a role for the mesenchymal stem cell (MSC) secretome in enhancing endometrial stromal cell motility. Abstract: Cyclic regeneration and repair of the endometrium are crucial for successful reproduction. Mesenchymal stem cells (MSCs) derived from bone marrow (BM-MSC) and umbilical cord (UC-MSC) facilitate tissue repair via their secretome, which contains growth factors and cytokines that promote wound healing. Despite the implication of MSCs in endometrial regeneration and repair, mechanisms remain unclear. This study tested the hypothesis that the BM-MSC and UC-MSC secretomes upregulate human endometrial stromal cell (HESC) proliferation, migration, and invasion and activate pathways to increase HESC motility. BM-MSCs were purchased from ATCC and cultured from the BM aspirate of three healthy female donors. UC-MSCs were cultured from umbilical cords of two healthy male term infants. Using indirect co-culture of MSCs and hTERT-immortalized HESCs via a transwell system, we demonstrated that co-culture of HESCs with BM-MSCs or UC-MSCs from all donors significantly increased HESC migration and invasion, whereas effects on HESC proliferation varied among BM-MSC and UC-MSC donors. Analysis of gene expression by mRNA sequencing and RT-qPCR showed that expression of CCL2 and HGF was upregulated in HESCs that had been cocultured with BM-MSCs or UC-MSCs. Validation studies revealed that exposure to recombinant CCL2 for 48 h significantly increased HESC migration and invasion. Increased HESC motility by the BM-MSC and UC-MSC secretome appears to be mediated in part by upregulated HESC CCL2 expression. Our data support the potential for leveraging MSC secretome as a novel cell-free therapy to treat disorders of endometrial regeneration.


Assuntos
Endométrio , Células-Tronco Mesenquimais , Secretoma , Células Estromais , Feminino , Humanos , Masculino , Diferenciação Celular , Movimento Celular/genética , Movimento Celular/fisiologia , Proliferação de Células , Técnicas de Cocultura , Endométrio/citologia , Endométrio/metabolismo , Células Epiteliais , Expressão Gênica , Células-Tronco Mesenquimais/metabolismo , Secretoma/metabolismo , Células Estromais/metabolismo , Células Estromais/fisiologia , Regulação para Cima , Células da Medula Óssea/fisiologia , Cordão Umbilical/citologia , Cordão Umbilical/fisiologia
3.
Front Endocrinol (Lausanne) ; 14: 1268990, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38344687

RESUMO

The endometrium is a resilient and highly dynamic tissue, undergoing cyclic renewal in preparation for embryo implantation. Cyclic endometrial regeneration depends on the intact function of several cell types, including parenchymal, endothelial, and immune cells, as well as adult stem cells that can arise from endometrial or extrauterine sources. The ability of the endometrium to undergo rapid, repeated regeneration without scarring is unique to this tissue. However, if this tissue renewal process is disrupted or dysfunctional, women may present clinically with infertility due to endometrial scarring or persistent atrophic/thin endometrium. Such disorders are rate-limiting in the treatment of female infertility and in the success of in vitro fertilization because of a dearth of treatment options specifically targeting the endometrium. A growing number of studies have explored the potential of adult stem cells, including mesenchymal stem cells (MSCs), to treat women with disorders of endometrial regeneration. MSCs are multipotent adult stem cells with capacity to differentiate into cells such as adipocytes, chondrocytes, and osteoblasts. In addition to their differentiation capacity, MSCs migrate toward injured sites where they secrete bioactive factors (e.g. cytokines, chemokines, growth factors, proteins and extracellular vesicles) to aid in tissue repair. These factors modulate biological processes critical for tissue regeneration, such as angiogenesis, cell migration and immunomodulation. The MSC secretome has therefore attracted significant attention for its therapeutic potential. In the uterus, studies utilizing rodent models and limited human trials have shown a potential benefit of MSCs and the MSC secretome in treatment of endometrial infertility. This review will explore the potential of MSCs to treat women with impaired endometrial receptivity due to a thin endometrium or endometrial scarring. We will provide context supporting leveraging MSCs for this purpose by including a review of mechanisms by which the MSC secretome promotes regeneration and repair of nonreproductive tissues.


Assuntos
Infertilidade Feminina , Células-Tronco Mesenquimais , Doenças Uterinas , Adulto , Feminino , Humanos , Cicatriz , Endométrio/patologia , Útero/metabolismo , Doenças Uterinas/metabolismo , Infertilidade Feminina/metabolismo
4.
F S Sci ; 3(3): 255-268, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35717521

RESUMO

OBJECTIVE: To determine whether bone marrow (BM)-derived cells engrafting the murine endometrium express the glucocorticoid receptor (GR) and androgen receptor (AR). Recent data demonstrate that BM is a long-term source of multiple hematopoietic and nonhematopoietic endometrial cell types. Important roles for glucocorticoids and androgens in regulating endometrial functions, including decidualization and early embryo attachment/invasion, have very recently emerged. Whether endometrial cells of BM origin express glucocorticoid or ARs has not been previously studied. DESIGN: Animal study. SETTING: Basic science laboratory. ANIMAL(S): Wild-type C57BL/6J male mice expressing enhanced green fluorescent protein (GFP) and syngeneic wild-type C57BL/6J female mice aged 6-9 weeks. INTERVENTION(S): Murine bone marrow transplant. MAIN OUTCOME MEASURE(S): Bone marrow cells were harvested from adult wild-type C57BL/6 mice and subjected to flow cytometry to identify the percentage of hematopoietic and nonhematopoietic cells expressing GR or AR. Uterine tissue sections from lethally irradiated syngeneic adult female C57BL/6 mice that had been recipients of BM transplants from adult male transgenic donor mice ubiquitously expressing GFP were studied. Immunohistochemistry was performed in the uterine tissue sections of the recipient mice at 5, 9, and 12 months after transplant using specific anti-GR, anti-AR, anti-GFP, anti-CD45 (pan leukocyte marker), and anti-F4/80 (murine macrophage marker) primary antibodies. Confocal laser microscopy was used to localize and quantitate BM-derived (GFP+) cell types in the endometrial stromal and epithelial compartments and determine whether BM-derived cell types in the murine endometrium express GR or AR. RESULT(S): Hematopoietic cells comprised 93.6%-96.6% of all cells in the BM, of which 98.1% ± 0.2% expressed GR and 92.2% ± 4.4% expressed AR. Nonhematopoietic cells comprised 0.4%-1.3% of BM, of which 52.8% ± 5.9% expressed GR and 48.9% ± 3.4% expressed AR. After BM transplant, the proportion of cells originating from BM in the endometrial stromal compartment increased over time, reaching 13.5% ± 2.3% at 12 months after transplant. In the epithelial compartments, <1% of the cells were of BM origin at 12 months after transplant. Most (60%-72%) GR+ and/or AR+ BM-derived cells in the stroma were hematopoietic (CD45+) cells, of which 37%-51% were macrophages. Nonetheless, 28%-33% of GR+ cells, and 28%-40% of AR+ BM-derived cells, were nonhematopoietic (CD45-) stromal cells of BM origin. CONCLUSION(S): A substantial number of BM-derived cells express GR and AR, suggesting a role for these cells in both glucocorticoid-regulated and androgen-regulated endometrial functions, such as proliferation and/or decidualization.


Assuntos
Endométrio , Células-Tronco Hematopoéticas , Receptores Androgênicos , Receptores de Glucocorticoides , Animais , Medula Óssea , Endométrio/citologia , Feminino , Células-Tronco Hematopoéticas/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Androgênicos/genética , Receptores de Glucocorticoides/genética
5.
Cells ; 11(9)2022 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-35563710

RESUMO

Ovarian stimulation is an indispensable part of IVF and is employed to produce multiple ovarian follicles. In women who undergo ovarian stimulation with gonadotropins, supraphysiological levels of estradiol, as well as a premature rise in progesterone levels, can be seen on the day of hCG administration. These alterations in hormone levels are associated with reduced embryo implantation and pregnancy rates in IVF cycles with a fresh embryo transfer. This article aims to improve the reader's understanding of the effects of elevated progesterone levels on human endometrial receptivity and oocyte/embryo quality. Based on current clinical data, it appears that the premature rise in progesterone levels exerts minimal or no effects on oocyte/embryo quality, while advancing the histological development of the secretory endometrium and displacing the window of implantation. These clinical findings strongly suggest that reduced implantation and pregnancy rates are the result of a negatively affected endometrium rather than poor oocyte/embryo quality. Understanding the potential negative impact of elevated progesterone levels on the endometrium is crucial to improving implantation rates following a fresh embryo transfer. Clinical studies conducted over the past three decades, many of which have been reviewed here, have greatly advanced our knowledge in this important area.


Assuntos
Endométrio , Progesterona , Implantação do Embrião/fisiologia , Transferência Embrionária , Endométrio/patologia , Feminino , Humanos , Oócitos , Gravidez , Progesterona/farmacologia
6.
Cells ; 11(4)2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-35203363

RESUMO

Endometrial cancer is the most common gynecological cancer, representing 3.5% of all new cancer cases in the United States. Abnormal stem cell-like cells, referred to as cancer stem cells (CSCs), reside in the endometrium and possess the capacity to self-renew and differentiate into cancer progenitors, leading to tumor progression. Herein we review the role of the endometrial microenvironment and sex hormone signaling in sustaining EC progenitors and potentially promoting dormancy, a cellular state characterized by cell cycle quiescence and resistance to conventional treatments. We offer perspective on mechanisms by which bone marrow-derived cells (BMDCs) within the endometrial microenvironment could promote endometrial CSC (eCSC) survival and/or dormancy. Our perspective relies on the well-established example of another sex hormone-driven cancer, breast cancer, in which the BM microenvironment plays a crucial role in acquisition of CSC phenotype and dormancy. Our previous studies demonstrate that BMDCs migrate to the endometrium and express sex hormone (estrogen and progesterone) receptors. Whether the BM is a source of eCSCs is unknown; alternatively, crosstalk between BMDCs and CSCs within the endometrial microenvironment could be an additional mechanism supporting eCSCs and tumorigenesis. Elucidating these mechanisms will provide avenues to develop novel therapeutic interventions for EC.


Assuntos
Neoplasias da Mama , Neoplasias do Endométrio , Medula Óssea/patologia , Neoplasias da Mama/patologia , Neoplasias do Endométrio/metabolismo , Endométrio , Feminino , Humanos , Células-Tronco Neoplásicas/patologia , Microambiente Tumoral
7.
Biol Reprod ; 106(5): 835-853, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35084017

RESUMO

Benign disorders of the human female reproductive system, such primary ovarian insufficiency and polycystic ovary syndrome are associated with infertility and recurrent miscarriage, as well as increased risk of adverse health outcomes, including cardiovascular disease and type 2 diabetes. For many of these conditions, the contributing molecular and cellular processes are poorly understood. The overarching similarities between mice and humans have rendered mouse models irreplaceable in understanding normal physiology and elucidating pathological processes that underlie disorders of the female reproductive system. The utilization of Cre-LoxP recombination technology, which allows for spatial and temporal control of gene expression, has identified the role of numerous genes in development of the female reproductive system and in processes, such as ovulation and endometrial decidualization, that are required for the establishment and maintenance of pregnancy in mammals. In this comprehensive review, we provide a detailed overview of Cre drivers with activity in the neuroendocrine-reproductive axis that have been used to study disruptions in key intracellular signaling pathways. We first summarize normal development of the hypothalamus, pituitary, ovary, and uterus, highlighting similarities and differences between mice and humans. We then describe human conditions resulting from abnormal development and/or function of the organ. Finally, we describe loss-of-function models for each Cre driver that elegantly recapitulate some key features of the human condition and are associated with impaired fertility. The examples we provide illustrate use of each Cre driver as a tool for elucidating genetic and molecular underpinnings of reproductive dysfunction.


Assuntos
Diabetes Mellitus Tipo 2 , Síndrome do Ovário Policístico , Animais , Feminino , Humanos , Integrases , Camundongos , Gravidez , Reprodução/genética
8.
Hum Reprod Update ; 25(1): 114-133, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30407544

RESUMO

BACKGROUND: The human uterine endometrium undergoes significant remodeling and regeneration on a rapid and repeated basis, after parturition, menstruation, and in some cases, injury. The ability of the adult endometrium to undergo cyclic regeneration and differentiation/decidualization is essential for successful human reproduction. Multiple key physiologic functions of the endometrium require the cells of this tissue to transition between mesenchymal and epithelial phenotypes, processes known as mesenchymal-epithelial transition (MET) and epithelial-mesenchymal transition (EMT). Although MET/EMT processes have been widely characterized in embryonic development and in the context of malignancy, mounting evidence demonstrates the importance of MET/EMT in allowing the endometrium the phenotypic and functional flexibility necessary for successful decidualization, regeneration/re-epithelialization and embryo implantation. OBJECTIVE AND RATIONALE: The objective of this review is to provide a comprehensive summary of the observations concerning MET and EMT and their regulation in physiologic uterine functions, specifically in the context of endometrial regeneration, decidualization and embryo implantation. SEARCH METHODS: Using variations of the search terms 'mesenchymal-epithelial transition', 'mesenchymal-epithelial transformation', 'epithelial-mesenchymal transition', 'epithelial-mesenchymal transformation', 'uterus', 'endometrial regeneration', 'endometrial decidualization', 'embryo implantation', a search of the published literature between 1970 and 2018 was conducted using the PubMed database. In addition, we searched the reference lists of all publications included in this review for additional relevant original studies. OUTCOMES: Multiple studies demonstrate that endometrial stromal cells contribute to the regeneration of both the stromal and epithelial cell compartments of the uterus, implicating a role for MET in mechanisms responsible for endometrial regeneration and re-epithelialization. During decidualization, endometrial stromal cells undergo morphologic and functional changes consistent with MET in order to accommodate embryo implantation. Under the influence of estradiol, progesterone and multiple other factors, endometrial stromal fibroblasts acquire epithelioid characteristics, such as expanded cytoplasm and rough endoplasmic reticulum required for greater secretory capacity, rounded nuclei, increased expression of junctional proteins which allow for increased cell-cell communication, and a reorganized actin cytoskeleton. During embryo implantation, in response to both maternal and embryonic-derived signals, the maternal luminal epithelium as well as the decidualized stromal cells acquire the mesenchymal characteristics of increased migration/motility, thus undergoing EMT in order to accommodate the invading trophoblast. WIDER IMPLICATIONS: Overall, the findings support important roles for MET/EMT in multiple endometrial functions required for successful reproduction. The endometrium may be considered a unique wound healing model, given its ability to repeatedly undergo repair without scarring or loss of function. Future studies to elucidate how MET/EMT mechanisms may contribute to scar-free endometrial repair will have considerable potential to advance studies of wound healing mechanisms in other tissues.


Assuntos
Endométrio/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Adulto , Movimento Celular , Implantação do Embrião/fisiologia , Feminino , Humanos , Menstruação/fisiologia , Gravidez , Células Estromais/fisiologia , Útero/metabolismo
9.
Am J Obstet Gynecol ; 215(5): 579.e1-579.e5, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27457114

RESUMO

Women with polycystic ovary syndrome have substantially higher rates of insulin resistance, impaired glucose tolerance, type 2 diabetes, dyslipidemia, and metabolic syndrome when compared with women without the disease. Given the high prevalence of these comorbidities, guidelines issued by the American College of Obstetricians and Gynecologists and the Endocrine Society recommend that all women with polycystic ovary syndrome undergo screening for impaired glucose tolerance and dyslipidemia with a 2 hour 75 g oral glucose tolerance test and fasting lipid profile upon diagnosis and also undergo repeat screening every 2-5 years and every 2 years, respectively. Although a hemoglobin A1C and/or fasting glucose are widely used screening tests for diabetes, both the American College of Obstetricians and Gynecologists and the Endocrine Society preferentially recommend the 2 hour oral glucose tolerance test in women with polycystic ovary syndrome as a superior indicator of impaired glucose tolerance/diabetes mellitus. However, we found that gynecologists underutilize current recommendations for metabolic screening in women with polycystic ovary syndrome. In an online survey study targeting American College of Obstetricians and Gynecologists fellows and junior fellows, 22.3% of respondents would not order any screening test at the initial visit for at least 50% of their patients with polycystic ovary syndrome. The most common tests used to screen for impaired glucose tolerance in women with polycystic ovary syndrome were hemoglobin A1C (51.0%) and fasting glucose (42.7%). Whereas 54.1% would order a fasting lipid profile in at least 50% of their polycystic ovary syndrome patients, only 7% of respondents order a 2 hour oral glucose tolerance test. We therefore call for increased efforts to encourage obstetrician-gynecologists to address metabolic abnormalities in their patients with polycystic ovary syndrome. Such efforts should include education of physicians early in their careers, at the medical student and resident level. Efforts should also include implementation of continuing medical education activities, both locally and at the national level, to improve understanding of the metabolic implications of polycystic ovary syndrome. Electronic medical record systems should be utilized to generate prompts for appropriate screening tests in patients with a diagnosis of polycystic ovary syndrome. Because obstetrician-gynecologists may be the only physicians seen by many polycystic ovary syndrome patients, particularly those in their young reproductive years, such interventions could effectively promote optimal preventative health care and early diagnosis of metabolic comorbidities in these at-risk women.


Assuntos
Dislipidemias/diagnóstico , Intolerância à Glucose/diagnóstico , Fidelidade a Diretrizes , Síndrome Metabólica/diagnóstico , Síndrome do Ovário Policístico/metabolismo , Glicemia/metabolismo , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/metabolismo , Dislipidemias/complicações , Dislipidemias/metabolismo , Feminino , Intolerância à Glucose/complicações , Intolerância à Glucose/metabolismo , Teste de Tolerância a Glucose/estatística & dados numéricos , Hemoglobinas Glicadas/metabolismo , Ginecologia , Humanos , Programas de Rastreamento , Síndrome Metabólica/complicações , Síndrome Metabólica/metabolismo , Obstetrícia , Síndrome do Ovário Policístico/complicações , Polissacarídeos/metabolismo , Guias de Prática Clínica como Assunto , Padrões de Prática Médica , Inquéritos e Questionários
10.
Artigo em Inglês | MEDLINE | ID: mdl-26796059

RESUMO

Uterine leiomyomas, or fibroids, are the most common benign tumor in reproductive aged women. Affected women may remain asymptomatic or may report symptoms related to abnormal uterine bleeding, infertility, or pelvic pain and pressure. Depending on a patient's symptomatology and reproductive plans, treatment options include expectant management, medical management (hormonal and non-hormonal), or surgical management (myomectomy or hysterectomy). In those wishing to defer surgical management, non-hormonal therapies such as non-steroidal anti-inflammatory drugs and tranexamic acid have been shown to decrease menstrual blood loss. In patients with more symptomatic leiomyomas, hormonal therapies such as gonadotropin-releasing hormone agonists and selective progesterone receptor modulators are effective at reducing leiomyoma volume, uterine size, and menstrual blood loss. This manuscript will detail the available and emerging hormonal and non-hormonal treatments for symptomatic uterine leiomyomas.


Assuntos
Anticoncepcionais Femininos/uso terapêutico , Hormônio Liberador de Gonadotropina/agonistas , Leiomioma/tratamento farmacológico , Neoplasias Uterinas/tratamento farmacológico , Antifibrinolíticos/uso terapêutico , Inibidores da Aromatase/uso terapêutico , Cabergolina , Colecalciferol/uso terapêutico , Anticoncepcionais Femininos/administração & dosagem , Anticoncepcionais Orais Hormonais/uso terapêutico , Anticoncepcionais Orais Sintéticos/uso terapêutico , Danazol/uso terapêutico , Agonistas de Dopamina/uso terapêutico , Ergolinas/uso terapêutico , Estrenos/uso terapêutico , Antagonistas de Estrogênios/uso terapêutico , Feminino , Gestrinone/uso terapêutico , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Humanos , Dispositivos Intrauterinos Medicados , Levanogestrel/administração & dosagem , Mifepristona/uso terapêutico , Norpregnadienos/uso terapêutico , Oximas/uso terapêutico , Planejamento de Assistência ao Paciente , Moduladores Seletivos de Receptor Estrogênico/uso terapêutico , Somatostatina/análogos & derivados , Ácido Tranexâmico/uso terapêutico , Vitaminas/uso terapêutico
11.
Menopause ; 22(12): 1285-8, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25988797

RESUMO

OBJECTIVE: This study aims to determine whether myometrial artery calcifications increase with age and whether uterine sections are an appropriate model for studying vascular aging. METHODS: An observational study of 172 women (aged 45 y or older) who underwent hysterectomy for benign indications at the University Hospital (Newark, NJ) between July 1, 2009 and June 1, 2012 was performed. Women with a history of malignancy, undocumented last menstrual period, or unavailable uterine tissue slides were excluded. H&E-stained uterine sections were evaluated for myometrial artery calcifications (defined as the presence of acellular densely basophilic material within the media of vessels) by a single pathologist in a blinded manner. RESULTS: Between July 1, 2009 and June 1, 2012, hysterectomies were performed on 441 women, 172 of whom met inclusion criteria. Seventeen women (9.9%) had myometrial artery calcifications detectable on H&E-stained tissue sections. None of 84 women aged 45 to 49 years, 2 of 51 women (3.9%) aged 50 to 59 years (aged 56 and 58 y), 10 of 27 women (37%) aged 60 to 69 years, and 5 of 10 women (50%) aged 70 to 81 years had myometrial artery calcifications. The prevalence of myometrial artery calcifications significantly increased with advancing age (P = 0.022). CONCLUSIONS: Myometrial artery calcifications increase with advancing age. Histological sections of uterine tissue from hysterectomy specimens seem to be a useful model for evaluating vascular aging markers.


Assuntos
Envelhecimento , Artérias , Miométrio/irrigação sanguínea , Calcificação Vascular/epidemiologia , Idoso , Artérias/patologia , Estudos Transversais , Feminino , Humanos , Histerectomia , Pessoa de Meia-Idade , Miométrio/patologia , Calcificação Vascular/patologia
12.
Womens Health (Lond) ; 11(1): 35-49, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25581054

RESUMO

Endometriosis is a complex gynecologic condition affecting 6-10% of reproductive aged women and is a major cause of chronic pain and infertility. Mechanisms of disease pathogenesis are poorly understood. Considerable evidence supports the existence of a stem cell population in the endometrium which provides a physiologic source of regenerative endometrial cells, and multiple lines of evidence now support a key role for stem cells in the pathogenesis of endometriosis. In addition, new blood vessel formation is critical for the establishment and maintenance of endometriotic implants, a process in which endothelial progenitor cells may play an integral role. These new insights into disease pathogenesis present exciting opportunities to develop targeted and more effective therapeutic options in the management of this common and challenging disease.


Assuntos
Endometriose/metabolismo , Endométrio/fisiologia , Regeneração/fisiologia , Células-Tronco/metabolismo , Endometriose/etiologia , Endométrio/citologia , Feminino , Humanos , Distúrbios Menstruais
13.
Biol Reprod ; 89(1): 7, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23699390

RESUMO

Human endometrium has the remarkable ability to regenerate all cellular compartments with every menstrual cycle; the cellular source remains unknown. The objective of the present study was to determine whether the bone marrow (BM) is a source of multiple endometrial cell types using a murine BM transplant model. BM cells were harvested from transgenic donor mice that ubiquitously express green fluorescent protein (GFP) and were injected into lethally irradiated, syngeneic female recipient mice. Recipients with successful hematopoietic reconstitution were sacrificed at 3, 5, 9, and 12 mo posttransplant, after which hysterectomy was performed. Numbers of GFP-positive, CD45-positive, and CD45-negative cells in the endometrial stromal and epithelial compartments were determined. In the stromal compartment, BM-derived cells (BMDCs) were detectable as early as 3 mo posttransplant, and the BM remained a long-term contributor of nonhematopoietic endometrial cells. Nonhematopoietic endometrial cells comprised 47.3%-72.2% of total BMDCs in the stromal compartment at 12 mo posttransplant. In contrast, BMDCs were not detected in the glandular or luminal epithelial compartments until 12 mo posttransplant. These data demonstrate that the BM is a significant source of nonhematopoietic endometrial stromal compartment cells and contributes to a much lesser extent to the epithelial compartments. That BM is a source of nonhematopoietic cells in the endometrial stromal and epithelial compartments provides a potential mechanism by which monthly regeneration of the endometrium may occur.


Assuntos
Células da Medula Óssea/fisiologia , Endométrio/citologia , Animais , Transplante de Medula Óssea , Células Epiteliais/citologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Animais
15.
Fertil Steril ; 90(6): 2068-72, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18249366

RESUMO

OBJECTIVE: To determine whether early measurement of the serum cytokines interleukin-2 receptor (IL-2R), IL-6, and IL-8 along with human chorionic gonadotropin (hCG) and progesterone (P(4)) can differentiate an ectopic from an intrauterine gestation. DESIGN: Retrospective analysis. SETTING: University-based fertility center. PATIENT(S): 75 women who underwent treatment with in vitro fertilization (IVF) and subsequently had an ectopic gestation (n = 15), spontaneous abortion (SAB) (n = 30), or term delivery (TD) (n = 30). INTERVENTION(S): Serum samples were obtained 14 (day 28) and 21 (day 35) days after oocyte retrieval. MAIN OUTCOME MEASURE(S): Serum concentrations of IL-2R, IL-6, IL-8, P(4), and hCG. RESULT(S): Median hCG readings on day 28 and day 35 were statistically significantly lower in the ectopic gestation group than in those with spontaneous abortion or term delivery. On day 28, median IL-8 levels were lower in the ectopic gestation group when compared with all intrauterine gestations combined. No statistically significant differences in IL-2R or IL-6 levels were noted between groups. Despite P(4) supplementation, median day-35 P(4) levels were lower in ectopic gestation than in the spontaneous abortion and term delivery cycles. CONCLUSION(S): In the setting of a rise or plateau in hCG levels, low day-28 IL-8 and day-35 P(4) levels suggested an extrauterine implantation. This assay combination may facilitate earlier diagnosis of an ectopic gestation when pregnancy location is unclear.


Assuntos
Implantação do Embrião , Fertilização in vitro , Interleucina-8/sangue , Gravidez Ectópica/sangue , Aborto Espontâneo/sangue , Adulto , Biomarcadores/sangue , Gonadotropina Coriônica/sangue , Feminino , Idade Gestacional , Humanos , Interleucina-6/sangue , Nascido Vivo , Valor Preditivo dos Testes , Gravidez , Resultado da Gravidez , Gravidez Ectópica/diagnóstico , Progesterona/sangue , Receptores de Interleucina-2/sangue , Estudos Retrospectivos , Fatores de Tempo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA