Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Stem Cells ; 36(6): 881-890, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29450941

RESUMO

Endometriosis is ectopic growth of endometrial tissue traditionally thought to arise through retrograde menstruation. We aimed to determine if cells derived from endometriosis could enter vascular circulation and lead to hematogenous dissemination. Experimental endometriosis was established by transplanting endometrial tissue from DsRed+ mice into the peritoneal cavity of DsRed- mice. Using flow cytometry, we identified DsRed+ cells in blood of animals with endometriosis. The circulating donor cells expressed CXCR4 and mesenchymal stem cell (MSC) biomarkers, but not hematopoietic stem cell markers. Nearly all the circulating endometrial stem cells originated from endometriosis rather than from the uterus. Cells expressing DsRed, CXCR4, and MSCs markers were identified in the peritoneal wall and surrounding vessels of recipient mice, contributing to both endometriosis and angiogenesis. Cells originating in endometriosis lesions migrated and implanted in lung tissue and displayed makers of differentiation, indicating retained multipotency. In vitro these cells demonstrated multipotency and were able to differentiate into adipogenic, osteogenic, and chondrogenic lineages. Endometriosis lesions also expressed high levels of CXCL12, the CXCR4 receptor ligand. Serum CXCL12 levels were greater than in sham control mice. In humans with endometriosis, serum CXCL12 levels were significantly higher than controls, suggesting that the CXCL12/CXCR4 axis is operational in women with spontaneous endometriosis as well. Stem cells, rather than differentiated cells from endometriosis, enter the circulation in response to CXCL12. We identify an endometriosis-derived stem cell population, a potential mechanism of dissemination of this disease and a potential target for treatment of endometriosis. Stem Cells 2018;36:881-890.


Assuntos
Endometriose/metabolismo , Células-Tronco Mesenquimais/metabolismo , Adulto , Animais , Diferenciação Celular , Modelos Animais de Doenças , Endometriose/patologia , Feminino , Humanos , Camundongos , Pessoa de Meia-Idade , Transdução de Sinais , Adulto Jovem
2.
Biol Reprod ; 90(6): 121, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24740602

RESUMO

Bazedoxifene (BZA), a selective estrogen receptor modulator (SERM), inhibits the action of estrogens on endometrial proliferation. Here, we evaluate the effect of a tissue-selective estrogen complex (TSEC) containing BZA and conjugated estrogens (CE) on ectopic endometrial lesions in a mouse model of endometriosis. Experimental endometriosis was created in 60 female CD-1 mice. The mice were randomly divided into 10 groups that received varying doses of either BZA (1, 2, 3, or 5 mg/kg/day), BZA (1, 2, 3, or 5 mg/kg/day) in combination with CE (3 mg/kg/day), CE treatment alone (3 mg/kg/day), or vehicle control for 8 wk. Treatment with BZA alone or the TSEC containing BZA/CE led to a decrease in endometriotic lesion size compared to controls. The mean surface area of the untreated lesions was 19.6 mm(2). Treatment with BZA or BZA/CE resulted in reduced lesion size (to 8.8 and 7.8 mm(2), respectively). No significant difference was found in lesion size between the BZA and BZA/CE treatment groups or between different doses of either treatment. Ovarian cyst formation was not evident in the treated groups. Treatment with the TSEC containing higher BZA dosages (3 and 5 mg/kg/day) led to significantly lower levels of estrogen receptor (Esr1) mRNA expression compared to the control treatment. No differences were observed in expression of progesterone receptor (Pgr). Immunohistochemical analysis also demonstrated a decrease in ESR protein. The combination of CE and BZA may prove to be a novel treatment option for endometriosis.


Assuntos
Endometriose/tratamento farmacológico , Estrogênios Conjugados (USP)/farmacologia , Indóis/farmacologia , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Endometriose/patologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Camundongos Endogâmicos , Tamanho do Órgão/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/patologia , Distribuição Aleatória , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Resultado do Tratamento
3.
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
4.
Am J Pathol ; 176(2): 1050-6, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20042667

RESUMO

Endometriosis is a major cause of chronic pain, infertility, medical and surgical interventions, and health care expenditures. Tissue factor (TF), the primary initiator of coagulation and a modulator of angiogenesis, is not normally expressed by the endothelium; however, prior studies have demonstrated that both blood vessels in solid tumors and choroidal tissue in macular degeneration express endothelial TF. The present study describes the anomalous expression of TF by endothelial cells in endometriotic lesions. The immunoconjugate molecule (Icon), which binds with high affinity and specificity to this aberrant endothelial TF, has been shown to induce a cytolytic immune response that eradicates tumor and choroidal blood vessels. Using an athymic mouse model of endometriosis, we now report that Icon largely destroys endometriotic implants by vascular disruption without apparent toxicity, reduced fertility, or subsequent teratogenic effects. Unlike antiangiogenic treatments that can only target developing angiogenesis, Icon eliminates pre-existing pathological vessels. Thus, Icon could serve as a novel, nontoxic, fertility-preserving, and effective treatment for endometriosis.


Assuntos
Endometriose/terapia , Imunoconjugados/farmacologia , Neovascularização Patológica/terapia , Doenças Peritoneais/terapia , Tromboplastina/antagonistas & inibidores , Tromboplastina/imunologia , Adulto , Animais , Células CHO , Cricetinae , Cricetulus , Sistemas de Liberação de Medicamentos , Endometriose/metabolismo , Endometriose/patologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Feminino , Humanos , Imunoconjugados/administração & dosagem , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Fragmentos Fc das Imunoglobulinas/farmacologia , Imunoterapia/métodos , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Neovascularização Patológica/metabolismo , Doenças Peritoneais/metabolismo , Doenças Peritoneais/patologia , Tromboplastina/metabolismo , Transplante Heterólogo
5.
Semin Thromb Hemost ; 37(2): 158-64, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21370218

RESUMO

Invasion of the decidua by extravillous trophoblasts (EVTs) is accompanied by thrombin generation from decidual cell (DC)-expressed tissue factor (TF). This TF protects against hemorrhage as EVTs breach capillaries and subsequently invade and remodel spiral arteries and arterioles. Pre-eclampsia (P-EC) is the world's leading cause of fetal and maternal morbidity and mortality. It is associated with decidual hemorrhage and maternal thrombophilias, which form excess thrombin from DCs, and with maternal infections and other inflammatory conditions that are associated with excess expression of the proinflammatory cytokines interleukin (IL)-1 ß and tumor necrosis factor (TNF) α. In human first-trimester leukocyte-free DCs, (1) thrombin enhances expression of soluble fms-like tyrosine kinase-1 (sFlt-1), a potent inhibitor of angiogenesis; (2) thrombin, IL-1ß and TNF-α increase monocyte-recruiting chemokine expression leading to a macrophage excess in the pre-eclamptic decidua. The pathogenesis of P-EC likely stems from shallow EVT invasion leading to impaired decidual vascular remodeling. The resulting reduced uteroplacental blood flow is associated with a hypoxic placenta, which appears to secrete excess sFlt-1 into the maternal plasma. A regulatory role for DCs in vascular remodeling is indicated because impaired decidual vascular remodeling could stem from an aberrant local antiangiogenic milieu elicited by excess sFlt-1 and/or macrophage-inhibited EVT decidual invasion.


Assuntos
Decídua/metabolismo , Pré-Eclâmpsia/patologia , Tromboplastina/metabolismo , Decídua/patologia , Feminino , Hemostasia/fisiologia , Humanos , Interleucina-1beta/metabolismo , Neovascularização Patológica , Pré-Eclâmpsia/sangue , Gravidez , Trombina/fisiologia , Trofoblastos/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/biossíntese
6.
Expert Rev Mol Med ; 12: e35, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21040617

RESUMO

Preterm birth (PTB) complicates more than 12% of all deliveries. Despite significant research, the aetiology of most cases of PTB remains elusive. Two major antecedents of PTB, intra-amniotic infection and decidual haemorrhage (abruption), can exhibit dissimilar demographic and genetic predispositions, despite sharing common molecular and cellular pathways. The use of high-throughput, high-dimensional technologies reveals substantial crosstalk between the coagulation and inflammation pathways. Tissue factor, thrombin and cytokines are key mediators of this crosstalk. Abruptions are associated with excess thrombin generated from decidual-cell-expressed tissue factor. Although thrombin is a primary mediator of the coagulation cascade, it can also promote inflammation-associated PTB by enhancing expression of matrix metalloproteinase and neutrophil-chemoattracting and -activating chemokines. Here, we provide novel insights into the molecular mechanisms and pathways leading to PTB in the setting of placental abruption.


Assuntos
Descolamento Prematuro da Placenta , Nascimento Prematuro/etiologia , Nascimento Prematuro/fisiopatologia , Feminino , Hemorragia/complicações , Humanos , Imuno-Histoquímica , Gravidez , Nascimento Prematuro/imunologia , Transdução de Sinais
7.
Am J Obstet Gynecol ; 203(4): 402.e1-4, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20708167

RESUMO

OBJECTIVE: Preimplantation factor is a novel embryo-derived peptide that influences key processes in early pregnancy implantation, including immunity, adhesion, remodeling, and apoptosis. Herein, we explore the effects of synthetic preimplantation factor on trophoblast invasion. STUDY DESIGN: Invasion patterns of immortalized cultured HTR-8 trophoblast cells were analyzed through Matrigel extracellular matrix ± synthetic preimplantation factor (25-100 nM) in a transwell assay. Effects were compared with epidermal growth factor 10 µg/mL, scrambled aminoacid sequence of preimplantation factor, or media alone as controls. RESULTS: Synthetic preimplantation factor enhances trophoblast invasion at physiologic doses (at 50 nM, 260%; 95% confidence interval [CI], 174-346%; P = .05; 100 nM ,178%; 95% CI, 170-184%; P < .02), compared with scrambled amnioacid sequence preimplantation factor or control media. Epidermal growth factor added to synthetic preimplantation factor does not further enhance trophoblast invasion (synthetic preimplantation factor 50 nM + epidermal growth factor, 238%; 95% CI, 237-239%; P < .03; synthetic preimplantation factor 100 nM + epidermal growth factor 269%; 95% CI, 265-273%; P < .04). CONCLUSION: Preimplantation factor should be further investigated as it shows a potential preventative or therapeutic role for pregnancy complications associated with inadequate trophoblast invasion.


Assuntos
Fatores Biológicos/administração & dosagem , Trofoblastos/efeitos dos fármacos , Movimento Celular , Células Cultivadas , Fator de Crescimento Epidérmico/administração & dosagem , Feminino , Humanos , Gravidez , Primeiro Trimestre da Gravidez , Trofoblastos/citologia
8.
Am J Obstet Gynecol ; 202(5): 459.e1-8, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20452489

RESUMO

OBJECTIVE: Preimplantation factor (PIF) is a novel, 15 amino acid peptide, secreted by viable embryos. This study aims to elucidate PIF's effects in human endometrial stromal cells (HESC) decidualized by estrogen and progestin, which mimics the preimplantation milieu, and in first-trimester decidua cultures (FTDC). STUDY DESIGN: HESC or FTDC were incubated with 100 nmol/L synthetic PIF or vehicle control. Global gene expression was analyzed using microarray and pathway analysis. Proteins were analyzed using quantitative mass spectrometry, and PIF binding by protein array. RESULTS: Gene and proteomic analysis demonstrate that PIF affects immune, adhesion, and apoptotic pathways. Significant up-regulation in HESC (fold change) include: nuclear factor-k-beta activation via interleukin-1 receptor-associated kinase binding protein 1 (53); Toll-like receptor 5 (9); FK506 binding protein 15, 133kDa protein (2.3); and Down syndrome cell adhesion molecule like 1 (16). B-cell lymphoma protein 2 was down-regulated in HESC (21.1) and FTDC (27.1). Protein array demonstrates PIF interaction with intracellular targets insulin-degrading enzyme and beta-K+ channels. CONCLUSION: PIF displays essential multitargeted effects, of regulating immunity, promoting embryo-decidual adhesion, and regulating adaptive apoptotic processes.


Assuntos
Decídua/citologia , Implantação do Embrião/fisiologia , Peptídeos/fisiologia , Gravidez/fisiologia , Células Cultivadas , Regulação para Baixo/fisiologia , Feminino , Humanos , Espectrometria de Massas , Análise Serial de Proteínas , Proteômica , Células Estromais/efeitos dos fármacos , Receptor 2 Toll-Like/metabolismo , Regulação para Cima/fisiologia
9.
Am J Pathol ; 172(6): 1571-9, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18467705

RESUMO

Preeclampsia, a common pregnancy disorder associated with an increase in systemic inflammation, is the leading cause of maternal and fetal morbidity and mortality throughout the world. It is associated with shallow extravillous trophoblast invasion of the decidua, leading to uteroplacental blood flow that is inadequate for the developing fetal-placental unit. In preeclamptic women, interleukin-6 (IL-6) levels in plasma, but not placenta, are elevated, prompting evaluation of the decidua as a potential source of this excess, circulating IL-6. The current study found significantly higher immunohistochemical staining for IL-6 in decidual cells from preeclamptic versus preterm, gestational age-matched control placentas. Pro-inflammatory cytokines associated with the genesis of preeclampsia (i.e., tumor necrosis factor-alpha and interleukin-1beta) enhanced IL-6 mRNA levels and increased secreted IL-6 levels in first trimester leukocyte-free decidual cell incubations, as measured by real time quantitative RT-PCR, ELISA, and Western blotting. Therefore, decidual cell-derived IL-6 may contribute to excess circulating IL-6 levels that can promote both endothelial cell dysfunction (and subsequent vascular dysfunction) and the pathogenesis of preeclampsia whereas locally elevated IL-6 levels may contribute to an excess of decidual macrophages implicated in shallow extravillous trophoblast invasion of the decidua.


Assuntos
Citocinas/biossíntese , Decídua/metabolismo , Interleucina-6/biossíntese , Pré-Eclâmpsia/metabolismo , Adulto , Células Cultivadas , Decídua/patologia , Feminino , Humanos , Interleucina-6/sangue , Pré-Eclâmpsia/patologia , Gravidez
10.
Oncotarget ; 10(23): 2282-2291, 2019 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-31040919

RESUMO

Endometriosis is an inflammatory gynecological disorder among reproductive-aged women caused by the engraftment and proliferation of endometrial cells outside the uterus, most commonly in the pelvis. It is thought that the disease arises primarily from retrograde menstruation where cells from the endometrium travel through the fallopian tubes to the peritoneal cavity. However, migration of endometriosis-derived cells to distant organs outside of the peritoneal cavity have not been explored. In the present study, we developed and validated a mouse model of disseminated endometriosis using syngeneic DsRed endometrial tissue introduced into the peritoneum of immunocompetent mice. Flow cytometry and immunofluorescence analysis, demonstrated the presence of endometriosis-derived cells in multiple organs (including lung, spleen, liver and brain) in the murine endometriosis model. Immunostaining revealed the presence of DsRed+/CD45- cells in brain, liver and lung. Engraftment occurred in all experimental animals examined. Cells from endometriotic lesions are capable of migration to and engraftment of multiple organs outside of the peritoneal cavity. Micrometastasis of endometriosis is a novel and frequent phenomenon. These data suggest that widespread dissemination of endometriosis may be common, clinically unrecognized and contribute to the diffuse clinical manifestations of this disease.

11.
Ann N Y Acad Sci ; 1127: 101-5, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18443336

RESUMO

Tissue factor (TF), is a cellular receptor that binds the ligand factor VII/VIIa to initiate the blood coagulation cascade. In addition to its role as the initiator of the hemostatic cascade, TF is known to be involved in angiogenesis via an interaction with factor VIIa and protease-activated receptor-2 (PAR-2). In this article we review previous studies from our laboratory demonstrating that the pattern and level of TF expression is altered in multiple cell types derived from eutopic and ectopic endometrium from women with endometriosis compared with normal endometrium. We posit that the inflammatory environment that occurs in ectopic and eutopic endometrium from patients with disease results in high TF expression that in turn, signals via PAR-2 to further produce inflammatory cytokine or chemokine production and macrophage recruitment. Thus, our studies suggest that TF might be an ideal target for therapeutic intervention in endometriosis.


Assuntos
Endometriose/diagnóstico , Endometriose/metabolismo , Endométrio/metabolismo , Regulação da Expressão Gênica , Neovascularização Patológica , Tromboplastina/metabolismo , Animais , Proliferação de Células , Citocinas/metabolismo , Feminino , Humanos , Imuno-Histoquímica/métodos , Infertilidade Feminina/metabolismo , Inflamação , Camundongos , Modelos Biológicos
12.
Ann N Y Acad Sci ; 1127: 67-72, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18443332

RESUMO

During extravascular trophoblast (EVT) invasion of the decidua, thrombin generated from decidual cell-expressed tissue factor (TF) forms a "hemostatic envelope" that protects against hemorrhage during the initial breaching of capillaries by EVTs and subsequent invasion and remodeling of the spiral arteries and arterioles. Preeclampsia, the world's leading cause of fetal and maternal morbidity and mortality, stems from shallow trophoblast invasion leading to incomplete vascular remodeling that impairs uteroplacental blood flow. A considerable subset of cases of preeclampsia is associated with decidual hemorrhage and maternal thrombophilias, which form excess thrombin from decidual cell-expressed TF. Thrombin affects several cell functions by binding to protease-activated receptors. In first-trimester decidual cells, thrombin enhances expression of sFlt-1, which can block the angiogenic effects of vascular endothelial growth factor (VEGF) and placental growth factor. By contrast, thrombin does not affect decidual cell VEGF expression. Thrombin-enhanced sFlt-1 expression by decidual cells, the predominant cell type encountered by invading cytotrophoblasts, could promote preeclampsia by interfering with angiogenesis-dependent vascular remodeling to reduce uteroplacental blood flow.


Assuntos
Decídua/metabolismo , Decídua/patologia , Hemostasia , Neovascularização Patológica , Pré-Eclâmpsia/diagnóstico , Tromboplastina/biossíntese , Animais , Feminino , Humanos , Camundongos , Modelos Biológicos , Gravidez , Complicações na Gravidez , Trombina/biossíntese , Trombina/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese
13.
Ann N Y Acad Sci ; 1127: 129-33, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18443340

RESUMO

Preeclampsia is associated with an increased release of factors from the placental syncytium into maternal blood, including the antiangiogenic factors soluble fms-like tyrosine kinase-1 and soluble endoglin, the antifibrinolytic factor plasminogen activator inhibitor-1, prostanoids, lipoperoxides, cytokines, and microparticles. These factors are suggested to promote maternal endothelium dysfunction and are associated with placental damage in pregnancies also complicated with intrauterine growth restriction (IUGR). In this report, we briefly describe the interaction of syncytial factors with hypoxia, reactive oxygen species, and apoptosis in the pathophysiology of preeclampsia and IUGR. Given the critical role of the syncytium in these complications of pregnancy, we also present a novel methodology in which laser capture microdissection followed by Western blotting is used to assess levels of syncytial Fas ligand, a key protein in the apoptotic cascade.


Assuntos
Retardo do Crescimento Fetal/patologia , Regulação da Expressão Gênica no Desenvolvimento , Células Gigantes/metabolismo , Neovascularização Fisiológica , Placenta/metabolismo , Pré-Eclâmpsia/patologia , Apoptose , Proteína Ligante Fas , Feminino , Retardo do Crescimento Fetal/diagnóstico , Humanos , Lasers , Modelos Biológicos , Pré-Eclâmpsia/diagnóstico , Gravidez , Espécies Reativas de Oxigênio , Trofoblastos/metabolismo
14.
Am J Reprod Immunol ; 80(5): e13028, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30106199

RESUMO

Initial studies on the chemokine stromal derived factor 1 (now referred to as CXCL12) were proposed to be enhanced in several diseases including those which affect the female reproductive tract. These include endometriosis, Asherman's syndrome, endometrial cancers, and ovarian cancers. Additionally, recent studies from our laboratory suggest that CXCL12 signaling is involved in leiomyomas (fibroids). These diseases present an inflammatory/hypoxic environment which further promotes pathology. At first, studies focused on signaling by CXCL12 via its well-known receptor, CXCR4. However, the discovery of CXCR7 as another receptor for CXCL12 with rather high binding affinity and recent reports about its involvement in endometrial disease and cancer progression has questioned the potential of "selective blockade"' of CXCR4 to treat these ailments. This review will focus on the signaling and effects of the potent chemokine CXCL12, and its long-known G protein-coupled receptor CXCR4, as well as the alternate receptor CXCR7 on the female reproductive tract and related diseases such as endometriosis, Asherman's syndrome, leiomyomas, endometrial cancer, and ovarian cancer. Although several other mechanisms are inherent to these diseases such as gene mutations, differential expression of miRNAs and epigenetics, for this review, we will focus on the CXCL12/CXCR4/CXCR7 axis as a novel target.


Assuntos
Endometriose/imunologia , Genitália Feminina/metabolismo , Ginatresia/imunologia , Neoplasias Ovarianas/imunologia , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Neoplasias Uterinas/imunologia , Animais , Feminino , Humanos , Terapia de Alvo Molecular , Transdução de Sinais
15.
Reprod Biol ; 18(1): 109-114, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29422377

RESUMO

Endometriosis is a common condition in reproductive-aged women characterized by ectopic endometrial lesions of varied appearance, including red, white, blue, black or powder burn coloration, which contribute to chronic pain and infertility. The immunoconjugate molecule (Icon) targets Tissue Factor, a transmembrane receptor for Factor VII/VIIa that is aberrantly expressed in the endothelium supporting ectopic endometrial tissue. Icon has been shown to cause regression of endometriosis in a murine model of disease but prior to this study had not been tested in non-human primates. This study evaluated Icon as a novel treatment for endometriosis in non-human primates (Papio anubis) using an adenoviral vector (AdIcon) delivery system. Female baboons (n = 15) underwent surgical induction of endometriosis. After laparoscopic confirmation of endometriosis lesions 6-weeks post-surgery, the treatment group (n = 7) received weekly intraperitoneal injections of viral particles carrying the sequence for Icon, resulting in expression of the protein, while the control group (n = 8) received no treatment. Icon preferentially reduced the number and volume of red vascularized lesions. Icon may present a novel treatment for endometriosis by degrading red vascularized lesions, likely by targeting tissue factor aberrantly expressed in the lesion vasculature.


Assuntos
Doenças dos Anexos/terapia , Endometriose/terapia , Fator VII/genética , Imunoconjugados/administração & dosagem , Fragmentos Fc das Imunoglobulinas/genética , Neovascularização Patológica/prevenção & controle , Proteínas Recombinantes de Fusão/genética , Tromboplastina/antagonistas & inibidores , Adenoviridae , Doenças dos Anexos/imunologia , Doenças dos Anexos/metabolismo , Doenças dos Anexos/patologia , Substituição de Aminoácidos , Animais , Endometriose/imunologia , Endometriose/metabolismo , Endometriose/patologia , Feminino , Terapia Genética , Vetores Genéticos , Humanos , Imunoconjugados/genética , Imunoglobulina G/genética , Terapia de Alvo Molecular , Mutação , Neovascularização Patológica/etiologia , Papio anubis , Pelve , Fragmentos de Peptídeos/genética , Distribuição Aleatória , Tromboplastina/metabolismo
16.
J Clin Endocrinol Metab ; 92(5): 1920-6, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17311863

RESUMO

CONTEXT: HOX genes are highly evolutionarily conserved regulators of embryonic development. HOXA10 also regulates differentiation of the adult reproductive tract and mammary gland in response to sex steroids. OBJECTIVE: We recently identified two HOXA10 estrogen response elements (EREs). Here we demonstrate that estrogen-responsive HOXA10 expression is cell type specific. DESIGN AND SETTING: We conducted an in vitro study at an academic medical center. MAIN OUTCOME MEASURE: Reporter assay, gel shift assays (electrophoretic mobility shift assay), and immunohistochemistry were done. RESULTS: The HOXA10 EREs and a specificity protein 1 (Sp1) binding site differentially drive the cell-type-specific E2 response. In electrophoretic mobility shift assays, both estrogen receptor-alpha and -beta bound both EREs but not the Sp1 site. In reporter assays, both EREs and the Sp1 site demonstrated estrogen responsiveness and tissue specificity; transiently transfected uterine Ishikawa cells or breast MCF-7 cells showed differential responses to E2 treatment. Each response element (Sp1, ERE1, and ERE2) drove distinct differential expression in each cell type. Sp1 protein was expressed in a menstrual-cycle stage-specific expression pattern in endometrium, first expressed in perivascular cells. CONCLUSIONS: Tissue specificity inherent to a regulatory element as well as differential cellular expression of transcription factors imparts differential tissue-specific estrogen responsiveness.


Assuntos
Estrogênios/farmacologia , Proteínas de Homeodomínio/genética , Fator de Transcrição Sp1/genética , Mama/citologia , Mama/efeitos dos fármacos , Mama/metabolismo , Células Cultivadas , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Genes Reporter/genética , Proteínas Homeobox A10 , Humanos , Imuno-Histoquímica , Luciferases/genética , Plasmídeos/genética , Receptores de Estrogênio/metabolismo , Elementos de Resposta/genética , Elementos de Resposta/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição Sp1/fisiologia , Transfecção , Útero/citologia , Útero/efeitos dos fármacos , Útero/metabolismo
17.
Thromb Haemost ; 97(2): 245-53, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17264954

RESUMO

Preeclampsia (PE), intrauterine growth restriction (IUGR) and abruption with or without fetal loss are associated with reduced uteroplacental blood flow, decidual vasculopathy, endothelial cell dysfunction, thrombosis, inflammation and hemorrhage. Our hypothesis is that reduced uteroplacental blood flow causes focal decidual hypoxia that generates vascular endothelial growth factor (VEGF). The latter acts directly on decidual endothelial cells to induce aberrant expression of tissue factor (TF), the primary initiator of coagulation. This in turn generates thrombin that induces: i) further TF expression; and ii) inflammatory cytokines. Both VEGF and TF induce aberrant angiogenesis-vessel maintenance reflected by endothelial cell fenestrations and induction of a prothrombotic surface causing both the decidual hemorrhage (i.e. abruption) and thrombosis (i.e. uteroplacental vascular insufficiency) observed in these adverse pregnancy outcomes. This novel hypothesis is supported by our finding of TF expression in decidual endothelium of pregnancies complicated by IUGR and/or fetal loss. Moreover, treatment of cultured endometrial endothelial cells with VEGF or thrombin induces TF protein and mRNA expression. Quantitative RT-PCR analysis indicates that thrombin enhances (>10-fold) the output of diverse inflammatory cytokines in these cultures. The greatest effect (>2-log) was seen on macrophage inflammatory protein 3alpha (MIP3alpha). In vitro, thrombin results in endometrial endothelial cell aggregations and changes in the apoptotic pathway. Thus, we postulate that reductions in uteroplacental flow initiate a cascade of molecular effects leading to hypoxia, thrombosis, inflammation, and endothelial cell dysfunction resulting in untoward pregnancy outcomes.


Assuntos
Endométrio/irrigação sanguínea , Células Endoteliais/metabolismo , Retardo do Crescimento Fetal/metabolismo , Circulação Placentária , Trombina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas Angiogênicas/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Agregação Celular , Células Cultivadas , Citocinas/metabolismo , Decídua/irrigação sanguínea , Células Endoteliais/efeitos dos fármacos , Feminino , Retardo do Crescimento Fetal/patologia , Retardo do Crescimento Fetal/fisiopatologia , Regulação da Expressão Gênica , Idade Gestacional , Humanos , NF-kappa B/metabolismo , Gravidez , Complicações na Gravidez/metabolismo , Complicações na Gravidez/fisiopatologia , RNA Mensageiro/metabolismo , Trombina/farmacologia , Tromboplastina/metabolismo , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/farmacologia
18.
Am J Obstet Gynecol ; 196(3): 268.e1-8, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17346550

RESUMO

OBJECTIVE: Excess decidual macrophage infiltration has been linked to preeclampsia and a failure of endovascular trophoblast invasion. Severe preeclampsia with shallow placentation has also been linked to acquired and inherited maternal thrombophilias and recurrent decidual hemorrhage, which generates thrombin from decidual cell-expressed tissue factor. Therefore, the current study evaluated whether thrombin affects monocyte chemoattractant protein-1 (MCP-1) expression in stromal cells that are derived from cycling and gestational endometrium. STUDY DESIGN: Stromal cells that are isolated from cycling endometrium and first trimester and term decidua were grown to confluence, treated for 7 days with 10(-8) mol/L estradiol (E2) + 10(-7) mol/L medroxyprogesterone acetate (MPA), then switched to a serum-free medium that contained the corresponding steroids +/- thrombin. MCP-1 protein release was measured by enzyme-linked immunosorbent assay and Western blot; MCP-1 messenger RNA levels were assessed by real-time quantitative reverse transcription-polymerase chain reaction. RESULTS: Secreted MCP-1 levels were not significantly different in stromal or decidual cell cultures that were incubated with E2 or with E2 + MPA. Thrombin increased immunoreactive MCP-1 expression in a dose-response fashion in first trimester and term decidual cells but not in endometrial stromal cells. Thrombin-induced MCP-1 protein output was unaffected by MPA but was abrogated by incubation with the thrombin inhibitor, hirudin. Unexpectedly, thrombin-enhanced MCP-1 protein expression was unaccompanied by corresponding changes in steady state MCP-1 messenger RNA levels, which suggests its effects were posttranslational. CONCLUSION: MCP-1 protein expression is up regulated by thrombin in decidual cells across gestation, but not in stromal cells from predecidualized cycling endometrium.


Assuntos
Quimiocina CCL2/biossíntese , Decídua/citologia , Decídua/metabolismo , Trombina/fisiologia , Células Cultivadas , Feminino , Humanos , Gravidez , Primeiro Trimestre da Gravidez , Nascimento a Termo
19.
Reprod Biol Endocrinol ; 4: 2, 2006 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-16445864

RESUMO

BACKGROUND: Neutrophils infiltrate the endometrium pre-menstrually and after long-term progestin only-contraceptive (LTPOC) treatment. Trafficking of neutrophils involves endothelial cell-expressed intercellular adhesion molecule (ICAM-1). Previous studies observed that ICAM-1 was immunolocalized to the endothelium of endometrial specimens across the menstrual cycle, but disagreed as to whether extra-endothelial cell types express ICAM-1 and whether ICAM-1 expression varies across the menstrual cycle. METHODS: Endometrial biopsies were obtained from women across the menstrual cycle and from those on LTPOC treatment (either Mirena or Norplant). The biopsies were formalin-fixed and paraffin-embedded with subsequent immunohistochemical staining for ICAM-1. RESULTS: The current study found prominent ICAM-1 staining in the endometrial endothelium that was of equivalent intensity in different blood vessel types irrespective of the steroidal or inflammatory endometrial milieu across the menstrual cycle and during LTPOC therapy. Unlike the endothelial cells, the glands were negative and the stromal cells were weakly positive for ICAM immunostaining. CONCLUSION: The results of the current study suggest that altered expression of ICAM-1 by endothelial cells does not account for the influx of neutrophils into the premenstrual and LTPOC-derived endometrium. Such neutrophil infiltration may depend on altered expression of neutrophil chemoattractants.


Assuntos
Anticoncepcionais Femininos/administração & dosagem , Endométrio/irrigação sanguínea , Endométrio/metabolismo , Endotélio Vascular/metabolismo , Molécula 1 de Adesão Intercelular/biossíntese , Levanogestrel/administração & dosagem , Adulto , Implantes de Medicamento , Feminino , Humanos , Imuno-Histoquímica , Ciclo Menstrual , Pessoa de Meia-Idade , Infiltração de Neutrófilos/efeitos dos fármacos
20.
Steroids ; 71(9): 768-75, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16806336

RESUMO

Tibolone and its metabolites were evaluated on matrix metalloproteinase (MMP) expression in human endometrial stromal cells (HESCs) under the hypothesis that these steroids would act as progestins on MMP-1, -2, and -3 expression. After 7 days of priming and 24h experimental incubation of confluent cultured HESCs, 10(-7) M medroxyprogesterone acetate (P) reduced MMP-1 to 49+/-34% (p<0.05) and MMP-3 to 33+/-22% of basal levels (mean+/-S.E.M., p<0.05, n=5). Although HESCs were unaffected by 10(-8) M estradiol (E), E+P reduced MMP-1 and MMP-3 levels an additional 2.5-fold from P alone. Tibolone and Delta-4 tibolone were equivalent to E+P in inhibiting MMP-1 and MMP-3 output, whereas 10(-6)M of 3alpha-OH or 3beta-OH tibolone was required to elicit significant inhibition of both MMPs (p<0.05). By contrast, none of the treatments affected HESC-secreted MMP-2 output. The ELISA results were confirmed by Western blotting and by substrate gel zymography. Quantitative RT-PCR demonstrated corresponding changes in MMP-1 and MMP-3 mRNA levels. Inhibition of MMP-1 and MMP-3 expression by tibolone and Delta-4 tibolone is consistent with the metabolism of tibolone to Delta-4 tibolone, and subsequent binding of Delta-4 tibolone to the progesterone receptor. Since 3alpha-OH and 3beta-OH tibolone bind exclusively to the estrogen receptor, their inhibition of MMP-1 and MMP-3 suggests metabolism by HESCs to Delta-4 tibolone. These observations help to explain the paradox that the endometrium becomes atrophic after tibolone administration despite the persistence in the circulation of 3alpha-OH and 3beta-OH tibolone, but not tibolone or Delta-4 tibolone.


Assuntos
Endométrio/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Metaloproteinases da Matriz/metabolismo , Norpregnenos/farmacologia , Progestinas/farmacologia , Células Estromais/efeitos dos fármacos , Células Estromais/enzimologia , Células Cultivadas , Feminino , Humanos , Metaloproteinases da Matriz/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA