Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Stem Cell Res Ther ; 15(1): 129, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38693588

RESUMO

BACKGROUND: Human bone marrow-derived stem cells (hBMDSCs) are well characterized mediators of tissue repair and regeneration. An increasing body of evidence indicates that these cells exert their therapeutic effects largely through their paracrine actions rather than clonal expansion and differentiation. Here we studied the role of microRNAs (miRNAs) present in extracellular vesicles (EVs) from hBMDSCs in tissue regeneration and cell differentiation targeting endometrial stromal fibroblasts (eSF). METHODS: Extracellular vesicles (EVs) are isolated from hBMDSCs, characterized by transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA) techniques. Extracted total RNA from EVs was subjected to RNA seq analysis. Transfection and decidualization studies were carried out in endometrial stromal fibroblasts (eSF). Gene expression was analyzed by qRTPCR. Unpaired t-test with Welch's correction was used for data analysis between two groups. RESULTS: We identified several microRNAs (miRNAs) that were highly expressed, including miR-21-5p, miR-100-5p, miR-143-3p and let7. MiR-21 is associated with several signaling pathways involved in tissue regeneration, quiescence, cellular senescence, and fibrosis. Both miR-100-5p and miR-143-3p promoted cell proliferation. MiR-100-5p specifically promoted regenerative processes by upregulating TGF-ß3, VEGFA, MMP7, and HGF. MiR-100-5p blocked differentiation or decidualization as evidenced by morphologic changes and downregulation of decidualization mediators including HOXA10, IGFBP1, PRL, PR-B, and PR. CONCLUSION: EVs delivered to tissues by hBMDSCs contain specific miRNAs that prevent terminal differentiation and drive repair and regeneration. Delivery of microRNAs is a novel treatment paradigm with the potential to replace BMDSCs in cell-free regenerative therapies.


Assuntos
Diferenciação Celular , Proliferação de Células , Endométrio , Exossomos , Fibroblastos , Células-Tronco Mesenquimais , MicroRNAs , Humanos , MicroRNAs/metabolismo , MicroRNAs/genética , Feminino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Exossomos/metabolismo , Endométrio/metabolismo , Endométrio/citologia , Fibroblastos/metabolismo , Fibroblastos/citologia , Regeneração/genética , Células da Medula Óssea/metabolismo , Células da Medula Óssea/citologia
2.
Stem Cells Int ; 2023: 1598127, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37545483

RESUMO

Endometriosis is a chronic inflammatory gynecological disorder regulated by estrogen and characterized by the growth of endometrial tissue outside the uterus. We have previously demonstrated that mesenchymal stem cells (MSCs) contribute directly to endometriosis. Here, we investigated an indirect effect; we hypothesized that MSCs may also impact the bone marrow (BM) by regulating bone marrow-derived inflammatory cells. Endometriosis was induced in mice by transplanting uterine tissue into recipient mice followed by BM transplant. Control or MSC conditioned BM was injected retro-orbitally. Direct administration of MSCs outside of the setting of BM conditioning did not alter endometriosis. Coculture of an undifferentiated macrophage cell line with MSCs in vitro led to a reduction of M1 and increased M2 macrophages as determined by fluorescence-activated cell sorting and western blot. Conditioning of BM with MSCs and transplantation into a mouse model inhibited endometriotic lesion development and reduced lesion volume by sevenfold compared to BM transplant without MSCs conditioning. Immunohistochemistry and immunofluorescence showed that MSC conditioned BM reduced the infiltration of macrophages and neutrophils into endometriotic lesions by twofold and decreased the proportion of M1 compared to M2 macrophages, reducing inflammation and likely promoting tissue repair. Expression of several inflammatory markers measured by quantitative real-time polymerase chain reaction, including tumor necrosis factor alpha and CXCR4, was decreased in the conditioned BM. Donor MSCs were not detected in recipient BM or endometriotic lesions, suggesting that MSCs actively program the transplanted BM. Taken together, these data show that individual characteristics of BM have an unexpected role in the development of endometriosis. BM remodeling and alterations in the inflammatory response are also potential treatments for endometriosis. Identification of the molecular basis for BM programing by MSCs will lead to a better understanding of the immune system contribution to this disease and may lead to new therapeutic targets for endometriosis.

3.
F S Sci ; 4(1): 65-73, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36252793

RESUMO

OBJECTIVE: To study the effect of intrauterine injection of C-X-C motif chemokine ligand 12 (CXCL12), also known as a stem cell chemoattractant (stromal cell-derived factor 1), on fertility and endometrial receptivity in mice with endometriosis. DESIGN: Laboratory study. SETTING: Academic Medical Center. ANIMAL(S): Fifty-six mice underwent chemotherapy and bone marrow transplantation. Thirty-six of these mice underwent either surgery to induce endometriosis (n = 20) or sham surgery (n = 16). INTERVENTION(S): Injection of CXCL12 as a potential therapeutic agent to improve fertility in endometriosis. MAIN OUTCOME MEASURE(S): Pregnancy rate, bone marrow-derived cell (BMDC) recruitment and endometrial receptivity markers. RESULT(S): The mice with or without endometriosis received a single uterine injection of either CXCL12 or placebo. Uterine injection of CXCL12 increased the pregnancy rates in a mouse model of endometriosis. Mice were euthanized after delivery, and implantation markers homeobox A11, alpha-v beta-3 integrin, and progesterone receptor were analyzed by immunohistochemistry, whereas green fluorescent protein positive BMDC recruitment was quantified by immunohistochemistry and immunofluorescence. The sham surgery groups without endometriosis had the highest cumulative pregnancy rate (100%) regardless of CXCL12 treatment. The endometriosis group treated with placebo had the lowest pregnancy rate. An increased pregnancy rate was noted in the endometriosis group after treatment with CXCL12. There was also an increase in BMDC recruitment and endometrial expression of progesterone receptor and alpha-v beta-3 integrin in the endometriosis group that received CXCL12 compared with that in the endometriosis group that received placebo. CONCLUSION(S): Uterine injection of CXCL12 increased the pregnancy rates in a mouse model of endometriosis. These results suggest that CXCL12 has a potential role as a therapeutic agent in women with infertility related to endometriosis and potentially other endometrial receptivity defects.


Assuntos
Endometriose , Infertilidade Feminina , Gravidez , Humanos , Feminino , Camundongos , Animais , Endometriose/tratamento farmacológico , Receptores de Progesterona , Ligantes , Integrinas , Quimiocinas
5.
F S Sci ; 3(2): 197-203, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35560017

RESUMO

OBJECTIVE: To evaluate whether Let-7b regulatory regions are methylated in endometriosis and whether there are specific CpG methylation sites that can be identified as key epigenetic regulatory locations. DESIGN: Laboratory study. SETTING: Academic Medical Center. PATIENT(S): Twenty-one women with (n = 12) and without (n = 9) endometriosis. INTERVENTION(S): Laboratory investigation. In vitro assessment of Let-7b methylation. MAIN OUTCOME MEASURE(S): Four targeted regions upstream of Let-7b predicted to be the regulatory regions of this microribonucleic acid (miRNA) were amplified using bisulfite-specific polymerase chain reaction. Deoxyribonucleic acid sequences were analyzed to determine methylation status at each predicted regulatory region and CpG island. RESULT(S): Regions were chosen on the basis of percent (%) GC content and data from Ensembl/ENCODE databases, which predict locations of promoters, enhancers, CTCF, and transcription factor binding sites as well as candidate cis-regulatory elements. A region 1,161 base pairs upstream of the Let-7b coding region was significantly differentially methylated in ectopic samples compared with eutopic endometrium from patients with endometriosis. Four specific CpG islands within this region 2 were further analyzed individually, and 1 was found to be significantly methylated in endometriosis. We identified that transcription factor SP1 was predicted to bind to a sequence that contained this specific methylated CpG in endometriosis. CONCLUSION(S): We identified differential Let-7b methylation in endometriosis, demonstrating that the epigenetic nature of the disease extends to the regulation of miRNAs. Methylation of this novel Let-7b regulatory region explains the decreased levels of this miRNA in endometriosis and is distinct from the regions implicated in regulating Let-7b in cancer. Understanding of the disease-specific mechanisms leading to diminished expression may allow for better understanding of the etiology of endometriosis as well as development of new treatment options.


Assuntos
Endometriose , MicroRNAs , Ilhas de CpG/genética , Endometriose/genética , Feminino , Humanos , Metilação , MicroRNAs/genética , Regiões Promotoras Genéticas , Fatores de Transcrição/genética
6.
Am J Obstet Gynecol ; 227(2): 248.e1-248.e8, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35351413

RESUMO

BACKGROUND: Epidemiologic studies have demonstrated an association between endometriosis and the subsequent development of cardiovascular disease. The direct effect of endometriosis on the progression of atherosclerotic, if any, has not been previously characterized. Endometriosis leads to systemic inflammation that could have consequences for cardiovascular health. Here, we reported the effects of endometriosis on the development of atherosclerosis in a murine model. OBJECTIVE: This study aimed to determine the contribution of endometriosis in promoting cardiovascular disease in a murine model of endometriosis. STUDY DESIGN: Endometriosis was induced in 18 apolipoprotein E-null mice, the standard murine model used to study atherosclerosis. Mice of the same strain were used as controls (n=18) and underwent sham surgery without inducing endometriosis. The formation of endometriotic lesions was confirmed after 25 weeks of induction. Atherosclerotic lesions were subjected to hematoxylin and eosin staining followed by measurement of the aortic root luminal area and wall thickness. The whole aorta was isolated, and Oil Red O staining was performed to quantify the lipid deposits or plaque formation; moreover, biochemical assays were carried out in serum to determine the levels of lipids and inflammatory-related cytokines. RESULTS: Apolipoprotein E mice with endometriosis exhibited increased aortic atherosclerosis compared with controls as measured using Oil Red O staining (7.9% vs 3.1%, respectively; P=.0004). Mice with endometriosis showed a significant 50% decrease in the aortic luminal area compared with sham mice (0.85 mm2 vs 1.46 mm2; P=.03) and a significant increase in aortic root wall thickness (0.22 mm vs 0.15 mm; P=.04). There was no difference in the lipoprotein profile (P<.05) between mice with endometriosis and sham mice. The serum levels of inflammatory cytokines interleukin 1 alpha, interleukin 6, interferon gamma, and vascular endothelial growth factor were significantly (P<.05)increased in the endometriosis mice. CONCLUSION: Our study used a murine model to determine the effect of endometriosis on atherosclerosis. Inflammation-related cytokines interleukin 1 alpha, interleukin 6, interferon gamma, and vascular endothelial growth factor (angiogenic factor) released by endometriotic lesions may contribute to the increased cardiovascular risks in women with endometriosis. To reduce the risk of cardiovascular disease, early identification and treatment of endometriosis are essential. Future treatments targeting inflammatory cytokines may help reduce the long-term risk of cardiovascular disease in women with endometriosis.


Assuntos
Aterosclerose , Doenças Cardiovasculares , Endometriose , Placa Aterosclerótica , Animais , Modelos Animais de Doenças , Feminino , Humanos , Inflamação/metabolismo , Interferon gama , Interleucina-1alfa , Interleucina-6 , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator A de Crescimento do Endotélio Vascular
7.
Stem Cell Res Ther ; 13(1): 107, 2022 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-35279204

RESUMO

BACKGROUND: Acute kidney injury (AKI) causes abrupt deterioration in kidney function that disrupts metabolic, electrolyte and fluid homeostasis. Although the prevalence of AKI is steadily increasing, no definitive treatment options are available, leading to severe morbidity and mortality. We evaluated the role of uterine-derived multipotent stem cells in kidney regeneration after ischemic AKI. METHODS: Female C57BL/6J mice were hysterectomized and subsequently subject to AKI by either unilateral or bilateral renal ischemia-reperfusion injury. Uterine-derived cells (UDCs), containing a population of uterine stem cells, were isolated from the uteri of female transgenic DsRed mice and injected intravenously to AKI mice. Engraftment of DsRed cells was analyzed by flow cytometry while serum creatinine levels were determined colorimetrically. Expression of UDC markers and cytokine markers were analyzed by immunohistochemical and qRT-PCR methods, respectively. The Kaplan-Meier method was used to analyze survival time while unpaired t test with Welch's correction used for data analysis between two groups. RESULTS: Mice with an intact uterus, and hence an endogenous source of UDCs, had a higher survival rate after bilateral ischemic AKI compared to hysterectomized mice. Mice treated with infusion of exogenous UDCs after hysterectomy/AKI had lower serum creatinine levels and higher survival rates compared to controls that did not receive UDCs. Engraftment of labeled UDCs was significantly higher in kidneys of bilateral ischemic AKI mice compared to those that underwent a sham surgery. When unilateral ischemic AKI was induced, higher numbers of UDCs were found in the injured than non-injured kidney. Immunofluorescence staining demonstrated double-positive DsRed/Lotus tetragonolobus agglutinin (LTA) positive cells and DsRed/CD31 positive cells indicating contribution of UDCs in renal tubular and vascular regeneration. Expression of Cxcl12, Bmp2, Bmp4, and Ctnf in renal tissue was significantly higher in the UDCs injection group than the control group. CONCLUSIONS: UDCs engrafted injured kidneys, contributed to proximal tubule and vascular regeneration, improved kidney function and increased survival in AKI mice. UDC administration is a promising new therapy for AKI. Endogenous uterine stem cells likely also preserve kidney function, suggesting a novel interaction between the uterus and kidney. We suggest that hysterectomy may have a detrimental effect on response to renal injury.


Assuntos
Injúria Renal Aguda , Traumatismo por Reperfusão , Injúria Renal Aguda/terapia , Animais , Feminino , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Regeneração , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/terapia , Células-Tronco/metabolismo , Útero/metabolismo
8.
J Cell Mol Med ; 26(9): 2566-2578, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35318804

RESUMO

Preterm birth is a major contributor to neonatal mortality and morbidity. Infection results in elevation of inflammation-related cytokines followed by infiltration of immune cells into gestational tissue. CXCL12 levels are elevated in preterm birth indicating it may have a role in preterm labour (PTL); however, the pathophysiological correlations between CXCL12/CXCR4 signalling and premature labour are poorly understood. In this study, PTL was induced using lipopolysaccharide (LPS) in a murine model. LPS induced CXCL12 RNA and protein levels significantly and specifically in myometrium compared with controls (3-fold and 3.5-fold respectively). Highest levels were found just before the start of labour. LPS also enhanced the infiltration of neutrophils, macrophages and T cells, and induced macrophage M1 polarization. In vitro studies showed that condition medium from LPS-treated primary smooth muscle cells (SMC) induced macrophage migration, M1 polarization and upregulated inflammation-related cytokines such as interleukin (IL)-1, IL-6 and tumor necrosis factor alpha (TNF-α). AMD3100 treatment in pregnant mice led to a significant decrease in the rate of PTL (70%), prolonged pregnancy duration and suppressed macrophage infiltration into gestation tissue by 2.5-fold. Further, in-vitro treatment of SMC by AMD3100 suppressed the macrophage migration, decreased polarization and downregulated IL-1, IL-6 and TNF-α expression. LPS treatment in pregnant mice induced PTL by increasing myometrial CXCL12, which recruits immune cells that in turn produce inflammation-related cytokines. These effects stimulated by LPS were completely reversed by AMD3100 through blocking of CXCL12/CXCR4 signalling. Thus, the CXCL12/CXCR4 axis presents an excellent target for preventing infection and inflammation-related PTL.


Assuntos
Trabalho de Parto Prematuro , Nascimento Prematuro , Animais , Quimiocina CXCL12/metabolismo , Citocinas/metabolismo , Feminino , Humanos , Inflamação/metabolismo , Interleucina-6/metabolismo , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Camundongos , Miométrio/metabolismo , Trabalho de Parto Prematuro/genética , Gravidez , Nascimento Prematuro/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
9.
Reprod Sci ; 29(8): 2382-2390, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-34515983

RESUMO

Regeneration of uterine tissue is an important physiological process that allows for maintenance of fertility after menstruation or pregnancy. Stem cells, especially bone marrow-derived progenitors, play a crucial role in this regeneration. Here, we describe the conversion of DsRed-labeled bone marrow-derived stem cells (BMDSCs) into specific uterine cell types with both differentiated and stem cell properties in a murine model. Irradiated recipient mice underwent bone marrow transplant with DsRed-expressing BMDSCs and were analyzed for engraftment and differentiation of BMDSCs in the uterus after 2, 6, and 16 weeks. Microarray and qRT-PCR analysis of bone marrow-derived cells obtained from the uterus identified upregulation of markers indicating a contribution to the population of stromal, epithelial, endothelial, and muscle cells, followed by a late expansion of epithelial cells. Other engrafted BMDSCs in the uterus were characterized by the continued expression of specific stem cell markers such as Sca1, CD44, CD146, and CD133, indicating the some BMDSCs remain as progenitor cells. BMDSCs established in recipient mice by the 16th week were sorted by flow cytometry using DsRed and progenitor cell surface markers. In vitro cell culture studies showed that single sorted cells had clonogenic properties. These results suggest that engrafted BMDSCs in the uterus had both a stem cell component and were able to differentiate into several differentiated cell types. The pool of progenitor cells likely continues to supply differentiated uterine cells in the process of uterine repair and remodeling.


Assuntos
Células da Medula Óssea , Fenômenos Fisiológicos , Animais , Medula Óssea , Células da Medula Óssea/metabolismo , Diferenciação Celular , Transdiferenciação Celular , Feminino , Camundongos , Gravidez , Células-Tronco/fisiologia , Útero
10.
Reprod Sci ; 29(1): 243-249, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34279849

RESUMO

Endometriosis is a gynecological disease affecting 6-10% of women of reproductive age. In addition to gynecologic symptoms, endometriosis is associated with various systemic effects, including inflammation, altered body weight, and behavioral changes. Previous murine studies demonstrate that endometriosis is causally inked to increased pain sensitization, behavioral changes, and low body mass index (BMI). One possible cellular target that may mediate some of these findings is the hypocretin/orexin neurons. This neuronal system plays a role in regulating wakefulness/sleep cycles, pain perception, and appetite. We hypothesize that endometriosis alters activity level of the hypocretin/orexin (Hcrt) neuronal system. Mice underwent endometriosis induction surgeries (endo) or sham surgeries (sham) for the development of the experimental model. Immunocytochemistry was performed on harvested samples from the lateral hypothalamus, and activation levels of Hcrt cells were examined by quantifying the expression of phosphorylation of cAMP-responsive element binding protein (CREB) in these cells after an acute stress in sham and endo mice. Mice with endometriosis had greater Hcrt neurons activation than sham mice. Mice with endometriosis fed with high fat diet showed a lower fat/body weight and fat/lean tissue ratio compared to mice without endometriosis. There was no significant difference in food intake between sham and endometriosis mice. These results demonstrate that endometriosis is associated with low body mass and increased hypocretin/orexin activity, which could be implicated in the behavioral changes and to differences in body composition.


Assuntos
Endometriose/metabolismo , Região Hipotalâmica Lateral/metabolismo , Neurônios/metabolismo , Orexinas/metabolismo , Animais , Peso Corporal/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Camundongos
11.
Mol Hum Reprod ; 27(4)2021 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-33693775

RESUMO

Endometriosis is a widespread gynecologic condition affecting up to 15% of women of reproductive age. The Janus kinase/signal transducer and activator of transcription (JAK/STAT3) pathway is upregulated in endometriosis and is a therapeutic target. Here we sought to determine the effect of Tofacitinib, a JAK inhibitor in widespread clinical use, on JAK/STAT signaling in endometriosis and lesion growth. Endometriosis was surgically induced in C57BL/6 mice using homologous uterine horn transplantation. Lesions were allowed to form over 4 weeks followed by Tofacitinib (10 mg/kg) or vehicle administered by oral gavage over 4 weeks. Tofacitinib treatment in vivo led to endometriosis lesion regression and reduced adhesion burden compared to vehicle treatment. In vitro studies on Ishikawa cells showed that Tofacitinib reduced hypoxia-inducible factor 1α and vascular endothelial growth factor mRNA levels at 12 and 24 h. Western blot analysis showed that Tofacitinib effectively reduced STAT3 phosphorylation in Ishikawa cells and human primary stromal and epithelial cells from eutopic endometrium of patients with and without endometriosis. This study suggests that the inhibition of JAK/STAT signaling using Tofacitinib may be a viable method for the treatment of endometriosis.


Assuntos
Endometriose , Animais , Endometriose/tratamento farmacológico , Endometriose/genética , Endometriose/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Piperidinas , Pirimidinas/farmacologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
Mol Cell Biochem ; 476(4): 1717-1727, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33428059

RESUMO

Endometriosis is an estrogen-dependent, inflammatory gynecological disorder characterized by the growth of endometrial cells in lesions outside the uterus. Bone marrow-derived cells (BMDCs) engraft lesions and increase lesion size. Do endometriosis cells regulate differentiation of engrafted BMDCs in the pathogenesis and growth of endometriosis? Here, we report endometriosis derived stromal cells promote the differentiation of BMDCs to stromal, epithelial and leukocyte cell fates through paracrine signaling. In-vitro studies demonstrated that both mRNA and protein levels of vimentin, cytokeratin and PD-1 were significantly increased in BMDCs cocultured with stromal cells from endometriosis (ENDO) patients compared to stromal cells from normal endometrium (CNTL). Increased expression of PD-1 has been reported in malignancy where it promotes T cell quiescence and immune tolerance. Increased PD-1 was also confirmed in-vivo where we showed that PD-1 expression was induced in BMDCs engrafted into endometriotic lesions in a murine model of endometriosis. AMD3100, an antagonist for CXCR4 receptor inhibited PD-1 expression in BMDCs suggesting that PD-1 induction requires CXCL12. These results suggest that endometriosis stimulated BMDC differentiation through paracrine signaling and increased T cell PD-1 expression. Increased PD-1 expression may be one mechanism by which endometriosis avoids immune surveillance.


Assuntos
Células da Medula Óssea/metabolismo , Diferenciação Celular , Endometriose/metabolismo , Regulação da Expressão Gênica , Células-Tronco Mesenquimais/metabolismo , Comunicação Parácrina , Receptor de Morte Celular Programada 1/biossíntese , Adolescente , Adulto , Células da Medula Óssea/patologia , Técnicas de Cocultura , Endometriose/patologia , Feminino , Humanos , Pessoa de Meia-Idade , Células Estromais/metabolismo , Células Estromais/patologia
13.
Reprod Sci ; 28(2): 426-434, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32812213

RESUMO

Endometriosis is an estrogen-dependent gynecological disorder that affects 10% of reproductive-aged women and causes pelvic pain and infertility. Bone marrow-derived stem cells (BMDCs) are known to engraft endometriosis in association with lesion growth; however, they do not undergo significant clonal expansion. The indirect effects of BMDCs on endometriosis growth and cell proliferation are not well characterized. Here, we demonstrate that BMDCs' co-culture increased endometrial stromal cell proliferation. In vitro studies using endometrial cells showed that BMDCs increased cell proliferation and activation of CDK1 in both an endometriosis cell line and primary endometrial stromal cells from women with endometriosis, however not in normal endometrial cells. In vivo studies using a mouse model of endometriosis showed increased CDK1+ expression associated with engrafted GFP + BMDCs. These results suggest that endometrial cell proliferation is induced by stem cell-derived trophic factors leading to the growth of endometriotic lesions. Targeting the specific signaling molecules secreted by BMDC may lead to novel therapeutic strategies for controlling cell proliferation in endometriosis.


Assuntos
Proliferação de Células , Endometriose/metabolismo , Endométrio/metabolismo , Células-Tronco Mesenquimais/metabolismo , Comunicação Parácrina , Células Estromais/metabolismo , Animais , Proteína Quinase CDC2/genética , Proteína Quinase CDC2/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Endometriose/genética , Endometriose/patologia , Endométrio/patologia , Feminino , Humanos , Transplante de Células-Tronco Mesenquimais , Camundongos Endogâmicos C57BL , Transdução de Sinais , Células Estromais/patologia
14.
Fertil Steril ; 113(6): 1224-1231.e1, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32416979

RESUMO

OBJECTIVE: To determine the relationship between steroid receptor expression and pain symptoms in endometriosis. DESIGN: Cross-sectional SETTING: University Hospital PATIENT(S): Women with endometriosis (N = 92). INTERVENTION(S): Tissue samples were obtained from patients with surgically diagnosed endometriosis. MAIN OUTCOME MEASURE(S): A tissue microarray (TMA) was generated from patients with endometriosis. Data were collected on the presence and severity of dysmenorrhea, deep dyspareunia, dyschezia, and nonmenstrual pain by use of a numerical rating scale (NRS) at the time of surgery and after 1 year. The intensity of receptor expression was evaluated through immunohistochemistry and measured according to an immunoreactive score (IRS). Clinical variables were correlated to IRS by multivariate logistic regression analysis. RESULTS: Estrogen receptor-α (ER-α), progesterone receptor (PR), androgen receptor (AR), and aromatase expression differed among study participants. ER-α expression was reduced by progestin therapy, whereas of expressions of PR, AR, and aromatase were unchanged. Higher ER-α expression increased the likelihood of moderate to severe dysmenorrhea and deep dyspareunia in women not receiving hormonal treatment. In women receiving progestin therapy, persistently higher ER-α expression was correlated with greater likelihood of deep dyspareunia, severe dyschezia, and endometriosis-associated pain persistence at 1 year. CONCLUSION(S): ER-α, PR, AR, and aromatase were all expressed in deep endometriosis. ER-α levels best correlated with severity of symptoms, which suggests that ER is a key driver of deep endometriosis. Progestin treatment was associated with a reduction of ER-α expression; however, failure of ER suppression by progestins was also a predictor of pain severity and recurrence at 1 year.


Assuntos
Constipação Intestinal/etiologia , Dismenorreia/etiologia , Dispareunia/etiologia , Endometriose/diagnóstico , Receptor alfa de Estrogênio/metabolismo , Adulto , Biomarcadores/metabolismo , Constipação Intestinal/diagnóstico , Constipação Intestinal/prevenção & controle , Estudos Transversais , Dismenorreia/diagnóstico , Dismenorreia/prevenção & controle , Dispareunia/diagnóstico , Dispareunia/prevenção & controle , Endometriose/complicações , Endometriose/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Medição da Dor , Valor Preditivo dos Testes , Progestinas/uso terapêutico , Recidiva , Medição de Risco , Fatores de Risco , Índice de Gravidade de Doença , Análise Serial de Tecidos , Resultado do Tratamento , Adulto Jovem
15.
Am J Obstet Gynecol ; 223(4): 557.e1-557.e11, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32165186

RESUMO

BACKGROUND: Endometriosis, a chronic disease that afflicts millions of women worldwide, has traditionally been diagnosed by laparoscopic surgery. This diagnostic barrier delays identification and treatment by years, resulting in prolonged pain and disease progression. Development of a noninvasive diagnostic test could significantly improve timely disease detection. We tested the feasibility of serum microRNAs as diagnostic biomarkers of endometriosis in women with gynecologic disease symptoms. OBJECTIVE: The objective of the study was to validate the use of a microRNA panel as a noninvasive diagnostic method for detecting endometriosis. STUDY DESIGN: This was a prospective study evaluating subjects with a clinical indication for gynecological surgery in an academic medical center. Serum samples were collected prior to surgery from 100 subjects. Women were selected based on the presence of symptoms, and laparoscopy was performed to determine the presence or absence of endometriosis. The control group was categorized based on absence of visual disease at the time of surgery. Circulating miRNAs, miR-125b-5p, miR-150-5p, miR-342-3p, miR-451a, miR-3613-5p, and let-7b, were measured in serum by quantitative real-time polymerase chain reaction in a blinded fashion without knowledge of disease status. Receiver-operating characteristic analysis was performed on individual microRNAs as well as combinations of microRNAs. An algorithm combining the expression values of these microRNAs, built using machine learning with a random forest classifier, was generated to predict the presence or absence of endometriosis on operative findings. This algorithm was then tested in an independent data set of 48 previously identified subjects not included in the training set (24 endometriosis and 24 controls) to validate its diagnostic performance. RESULTS: The mean age of women in the study population was 34.1 and 36.9 years for the endometriosis and control groups, respectively. Control group subjects displayed varying pathologies, with leiomyoma occurring the most often (n = 39). Subjects with endometriosis had significantly higher expression levels of 4 serum microRNAs: miR-125b-5p, miR-150-5p, miR-342-3p, and miR-451a. Two serum microRNAs showed significantly lower levels in the endometriosis group: miR-3613-5p and let-7b. Individual microRNAs had receiver-operating characteristic areas under the curve ranging from 0.68 to 0.92. A classifier combining these microRNAs yielded an area under the curve of 0.94 when validated in the independent set of subjects not included in the training set. Analysis of the expression levels of each microRNA based on revised American Society of Reproductive Medicine staging revealed that all microRNAs could distinguish stage I/II from control and stage III/IV from control but that the difference between stage I/II and stage III/IV was not significant. Subgroup analysis revealed that neither phase of the menstrual cycle or use of hormonal medication had a significant impact on the expression levels in the microRNAs used in our algorithm. CONCLUSION: This is the first report showing that microRNA biomarkers can reliably differentiate between endometriosis and other gynecological pathologies with an area under the curve >0.9 across 2 independent studies. We validated the performance of an algorithm based on previously identified microRNA biomarkers, demonstrating their potential to detect endometriosis in a clinical setting, allowing earlier identification and treatment. The ability to diagnose endometriosis noninvasively could reduce the time to diagnosis, surgical risk, years of discomfort, disease progression, associated comorbidities, and health care costs.


Assuntos
Endometriose/diagnóstico , MicroRNAs/sangue , Adulto , Algoritmos , Área Sob a Curva , Endometriose/sangue , Estudos de Viabilidade , Feminino , Humanos , Laparoscopia , Aprendizado de Máquina , Pessoa de Meia-Idade , Estudos Prospectivos , Curva ROC , Índice de Gravidade de Doença
16.
Reprod Sci ; 27(9): 1724-1730, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32020550

RESUMO

Uterine leiomyomas, also known as fibroids or myomas, are a common benign gynecologic tumor found in women of reproductive age. Though advances have been made in understanding leiomyomas, the etiology and pathogenesis of this disease are not fully characterized. Current evidence supports a role of putative human uterine stem/progenitor cells in the onset of uterine disease such as uterine myomas. In this study, we report that increased expression of CXCL12 in leiomyomas recruits bone marrow-derived cells (BMDCs) that may contribute to leiomyoma growth. Tissue was collected from leiomyomas or control myometrium from women with or without leiomyomas. qRT-PCR analysis showed increased expression of CXCL12 and decreased CXCR4 expression in the leiomyoma and myometrium of women with leiomyoma compared with normal myometrium. Increased CXCL12 protein secretion from cultured myoma cells was confirmed by ELISA. Further, we found that BMDCs migration was increased toward leiomyoma conditioned medium compared with conditioned medium from normal myometrium. CXCR4 antagonist AMD3100 completely blocked this migration. Engraftment of BMDCs significantly increased in myoma of mouse uteri treated with CXCL12 compared with placebo. We conclude that CXCL12 may play a role in leiomyomas growth by attracting bone marrow-derived cells to leiomyoma. Therefore, CXCL12 and its receptors are novel targets for leiomyoma therapy.


Assuntos
Células da Medula Óssea/metabolismo , Movimento Celular/fisiologia , Quimiocina CXCL12/metabolismo , Leiomioma/metabolismo , Neoplasias Uterinas/metabolismo , Útero/metabolismo , Adulto , Animais , Benzilaminas , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/patologia , Movimento Celular/efeitos dos fármacos , Quimiocina CXCL12/antagonistas & inibidores , Ciclamos , Feminino , Compostos Heterocíclicos/farmacologia , Humanos , Leiomioma/patologia , Camundongos , Pessoa de Meia-Idade , Neoplasias Uterinas/patologia , Útero/efeitos dos fármacos , Útero/patologia
17.
J Cell Mol Med ; 24(4): 2464-2474, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31904910

RESUMO

Adult stem cells have a major role in endometrial physiology, including remodelling and repair. However, they also have a critical role in the development and progression of endometriosis. Bone marrow-derived stem cells engraft eutopic endometrium and endometriotic lesions, differentiating to both stromal and epithelial cell fates. Using a mouse bone marrow transplantation model, we show that bone marrow-derived cells engrafting endometriosis express CXCR4 and CXCR7. Targeting either receptor by the administration of small molecule receptor antagonists AMD3100 or CCX771, respectively, reduced BM-derived stem cell recruitment into endometriosis implants. Endometriosis lesion size was decreased compared to vehicle controls after treatment with each antagonist in both an early growth and established lesion treatment model. Endometriosis lesion size was not effected when the local effects of CXCL12 were abrogated using uterine-specific CXCL12 null mice, suggesting an effect primarily on bone marrow cell migration rather than a direct endometrial effect. Antagonist treatment also decreased hallmarks of endometriosis physiopathology such as pro-inflammatory cytokine production and vascularization. CXCR4 and CXCR7 antagonists are potential novel, non-hormonal therapies for endometriosis.


Assuntos
Benzilaminas/farmacologia , Células da Medula Óssea/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Ciclamos/farmacologia , Endometriose/tratamento farmacológico , Endométrio/efeitos dos fármacos , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR/antagonistas & inibidores , Células-Tronco Adultas/efeitos dos fármacos , Células-Tronco Adultas/metabolismo , Animais , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea/métodos , Endometriose/metabolismo , Endométrio/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/efeitos dos fármacos , Útero/efeitos dos fármacos , Útero/metabolismo
18.
Reprod Sci ; 26(12): 1633-1639, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31530098

RESUMO

Primary ovarian insufficiency (POI) is defined as ovarian dysfunction in women younger than 40 years. It affects 1% of the women in this age-group and can occur iatrogenically after chemotherapy. Stem cells have been used in attempt to restore ovarian function in POI. In particular, endometrial mesenchymal stem cells (eMSCs) are easily obtainable in humans and have shown great potential for regenerative medicine. Here, we studied the potential for uterine cell (UC) suspensions containing eMSCs to improve ovarian function in a murine model of chemotherapy-induced POI. Green fluorescent protein (GFP)-labeled UC or phosphate-buffered solution (PBS) was delivered intravenously after chemotherapy. There was a significant increase in oocytes production and serum anti-Müllerian hormone concentrations after 6 weeks, as well as a 19% higher body mass in UC-treated mice. Similarly, we observed an increased number of pups in mice treated with UC than in mice treated with PBS. None of the oocytes or pups incorporated GFP, suggesting that there was no contribution of these stem cells to the oocyte pool. We conclude that treatment with UC indirectly improved ovarian function in mice with chemotherapy-induced POI. Furthermore, our study suggests that endometrial stem cell therapy may be beneficial to young women who undergo ovotoxic chemotherapy.


Assuntos
Endométrio/citologia , Transplante de Células-Tronco Mesenquimais , Ovário/metabolismo , Insuficiência Ovariana Primária/terapia , Animais , Hormônio Antimülleriano/sangue , Modelos Animais de Doenças , Endométrio/metabolismo , Feminino , Camundongos , Insuficiência Ovariana Primária/metabolismo
19.
PLoS Biol ; 17(9): e3000421, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31513564

RESUMO

Decidua is a transient uterine tissue shared by mammals with hemochorial placenta and is essential for pregnancy. The decidua is infiltrated by many immune cells promoting pregnancy. Adult bone marrow (BM)-derived cells (BMDCs) differentiate into rare populations of nonhematopoietic endometrial cells in the uterus. However, whether adult BMDCs become nonhematopoietic decidual cells and contribute functionally to pregnancy is unknown. Here, we show that pregnancy mobilizes mesenchymal stem cells (MSCs) to the circulation and that pregnancy induces considerable adult BMDCs recruitment to decidua, where some differentiate into nonhematopoietic prolactin-expressing decidual cells. To explore the functional importance of nonhematopoietic BMDCs to pregnancy, we used Homeobox a11 (Hoxa11)-deficient mice, having endometrial stromal-specific defects precluding decidualization and successful pregnancy. Hoxa11 expression in BM is restricted to nonhematopoietic cells. BM transplant (BMT) from wild-type (WT) to Hoxa11-/- mice results in stromal expansion, gland formation, and marked decidualization otherwise absent in Hoxa11-/- mice. Moreover, in Hoxa11+/- mice, which have increased pregnancy losses, BMT from WT donors leads to normalized uterine expression of numerous decidualization-related genes and rescue of pregnancy loss. Collectively, these findings reveal that adult BMDCs have a previously unrecognized nonhematopoietic physiologic contribution to decidual stroma, thereby playing important roles in decidualization and pregnancy.


Assuntos
Células da Medula Óssea/fisiologia , Decídua/citologia , Implantação do Embrião , Células-Tronco Mesenquimais/fisiologia , Gravidez/fisiologia , Animais , Feminino , Proteínas de Homeodomínio/genética , Masculino , Camundongos Knockout
20.
Cell Death Dis ; 10(8): 592, 2019 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-31391459

RESUMO

Since publication of this article, Dr Ramanaiah Mamillapalli reported that his last name had published incorrectly as Ramillapalli. The publisher apologizes to the authors and to readers for this error, which has not been fixed in the original article.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...