Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Reprod Biomed Online ; 47(1): 35-50, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37142478

RESUMO

RESEARCH QUESTION: Advanced glycation end-products (AGE) are elevated in the uterine environment of obese infertile women. Can the detrimental effects of AGE on endometrial epithelial cells be mitigated with therapeutics, and recapitulated in a more physiologically relevant primary model (organoids)? DESIGN: Human endometrial epithelial cells (ECC-1) were exposed to AGE at concentrations physiologically representative of uterine fluid in lean or obese individuals, and three potential therapeutics: 25 nmol/l receptor for AGE (RAGE) antagonist FPS-ZM1, 100 µmol/l metformin, or a combination of antioxidants (10 µmol/l N-acetyl-l-cysteine, 10 µmol/l N-acetyl-l-carnitine and 5 µmol/l α-lipoic acid). Real-time cell analysis (xCELLigence, ACEA Biosciences) determined the rate of adhesion and proliferation. The proliferation of organoid-derived cells and secretion of cytokines from organoids was characterized in the presence of AGE (n = 5). The uterine fluid of women undergoing assisted reproduction was profiled for AGE-associated inflammatory markers (n = 77). RESULTS: ECC-1 proliferation was reduced by AGE from obese versus lean conditions and vehicle control (P = 0.04 and P < 0.001, respectively), and restored to a proliferation corresponding to lean conditions by antioxidants. AGE influenced organoid derived primary endometrial epithelial cell proliferation in a donor-dependent manner. AGE increased the organoid secretion of the proinflammatory cytokine CXCL16 (P = 0.006). Clinically, CXCL16 correlated positively to maternal body mass index (R = 0.264, P = 0.021) and intrauterine glucose concentration (R = 0.736, P < 0.0001). CONCLUSIONS: Physiologically relevant concentrations of AGE alter endometrial epithelial cell function. Antioxidants restore the rate of proliferation of AGE-treated endometrial epithelial (ECC-1) cells. Primary endometrial epithelial cells, cultured as organoids, demonstrate altered proliferation and CXCL16 secretion in the presence of AGE equimolar with the uterine fluid from obese individuals.


Assuntos
Infertilidade Feminina , Doenças Uterinas , Feminino , Humanos , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Infertilidade Feminina/metabolismo , Reação de Maillard , Endométrio/metabolismo , Proliferação de Células , Obesidade/complicações , Obesidade/metabolismo , Receptor para Produtos Finais de Glicação Avançada
2.
Reprod Biomed Online ; 44(1): 14-26, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34810095

RESUMO

RESEARCH QUESTION: Is the blastocyst's idiosyncratic metabolic production of lactate, and creation of a specialized microenvironment at the implatation site, an important mediator of maternal-fetal signalling to promote endometrial receptivity and implantation? DESIGN: Hormonally primed ECC-1 and Ishikawa cells were used to assess functional changes to the endometrial epithelium after exposure to lactic acid (LA), LA with neutralized pH (nLA) or acidic pH (pHL). Tight junction integrity (transepithelial resistance [TER]), cellular proliferation or changes to gene expression by RT-PCR were analysed. The effect of LA on Endometrial stromal cells decidualization and migratory capacity, and HUVEC endothelial tube formation and angiogenesis, were also assessed. RESULTS: Treatment of ECC-1 cells with 2.5 mM (P = 0.0037), 5 mM (P = 0.0044), 7.5 mM and 10 mM (P = 0.003) (P = 0.0021) LA significantly decreased the rate of cellular proliferation while TER was decreased with exposure to 2.5 mM LA (P = 0.024), 5 mM LA (P = 0.021) and 7.5 mM LA (P = 0.033). Exposure to nLA or pHL had no effect on proliferation or TER. Upregulation of GLUT4 (P = 0.002), GPR81 (P = 0.048), VEGF, SNAI1 (both P < 0.001) and RELA (P = 0.023) mRNA expression was observed after exposure of Ishikawa cells to combined LA plus pHL. Lactic acid increased the migratory capacity of decidualized stromal cells (P = 0.047) without changing the extent of decidualization. HUVEC tube formation was significantly increased by 5 mM LA exposure (P = 0.009). CONCLUSIONS: The identification of LA as an important mediator in the maternal-fetal dialogue underpinning implantation is supported. Further examination of the role of LA within the infertile or compromised endometrium could improve natural and assisted pregnancy success and needs further investigation.


Assuntos
Implantação do Embrião , Ácido Láctico , Blastocisto/metabolismo , Implantação do Embrião/fisiologia , Endométrio/metabolismo , Feminino , Humanos , Concentração de Íons de Hidrogênio , Ácido Láctico/metabolismo , Gravidez , Células Estromais/metabolismo
3.
Mol Hum Reprod ; 27(10)2021 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-34524461

RESUMO

Adequate endometrial stromal cell (ESC) decidualization is vital for endometrial health. Given the importance of extracellular vesicles (EVs) in intercellular communication, we investigated how their protein landscape is reprogrammed and dysregulated during decidual response. Small EVs (sEVs) from human ESC-conditioned media at Day-2 and -14 following decidual stimuli were grouped as well- (WD) or poorly decidualized (PD) based on their prolactin secretion and subjected to mass spectrometry-based quantitative proteomics. On Day 2, in PD- versus WD-ESC-sEVs, 17 sEV- proteins were down-regulated (C5, C6; complement/coagulation cascades, and SERPING1, HRG; platelet degranulation and fibrinolysis) and 39 up-regulated (FLNA, COL1A1; focal adhesion, ENO1, PKM; glycolysis/gluconeogenesis, and RAP1B, MSN; leukocyte transendothelial migration). On Day 14, in PD- versus WD-ESC-sEVs, FLNA was down-regulated while 21 proteins were up-regulated involved in complement/coagulation cascades (C3, C6), platelet degranulation (SERPINA4, ITIH4), B-cell receptor signalling and innate immune response (immunoglobulins). Changes from Days 2 to 14 suggested a subsequent response in PD-ESC-sEVs with 89 differentially expressed proteins mostly involved in complement and coagulation cascades (C3, C6, C5), but no change in WD-ESC-sEVs ESC. Poor decidualization was also associated with loss of crucial sEV-proteins for cell adhesion and invasion (ITGA5, PFN1), glycolysis (ALDOA, PGK1) and cytoskeletal reorganization (VCL, RAC1). Overall, this study indicates varied ESC response even prior to decidualization and provides insight into sEVs-proteomes as a benchmark of well-decidualized ESC. It shows distinct variation in sEV-protein composition depending on the ESC decidual response that is critical for embryo implantation, enabling and limiting trophoblast invasion during placentation and sensing a healthy embryo.


Assuntos
Endométrio/metabolismo , Vesículas Extracelulares/metabolismo , Fibroblastos/metabolismo , Proteoma , Células Estromais/metabolismo , Adulto , Células Cultivadas , Decídua/metabolismo , Implantação do Embrião , Endométrio/efeitos dos fármacos , Endométrio/ultraestrutura , Estradiol/farmacologia , Vesículas Extracelulares/efeitos dos fármacos , Vesículas Extracelulares/ultraestrutura , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/ultraestrutura , Humanos , Acetato de Medroxiprogesterona/farmacologia , Placentação , Gravidez , Proteômica , Células Estromais/efeitos dos fármacos , Células Estromais/ultraestrutura , Fatores de Tempo , Adulto Jovem
4.
Proteomics ; 20(1): e1900250, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31825151

RESUMO

In vitro fertilization has overcome infertility issues for many couples. However, achieving implantation of a viable embryo into the maternal endometrium remains a limiting step in optimizing pregnancy success. The molecular mechanisms which characterize the transient state of endometrial receptivity, critical in enabling embryo-endometrial interactions, and proteins which underpin adhesion at the implantation interface, are limited in humans despite these temporally regulated processes fundamental to life. Hence, failure of implantation remains the "final frontier" in infertility. A human coculture model is utilized utilizing spheroids of a trophectoderm (trophoblast stem) cell line, derived from pre-implantation human embryos, and primary human endometrial epithelial cells, to functionally identify "fertile" versus "infertile" endometrial epithelium based on adhesion between these cell types. Quantitative proteomics identified proteins associated with human endometrial epithelial receptivity ("epithelial receptome") and trophectoderm adhesion ("adhesome"). As validation, key "epithelial receptome" proteins (MAGT-1/CDA/LGMN/KYNU/PC4) localized to the epithelium of receptive phase (mid-secretory) endometrium obtained from fertile, normally cycling women but is largely absent from non-receptive (proliferative) phase tissues. Factors involved in embryo-epithelium interaction in successive temporal stages of endometrial receptivity and implantation are demonstrated and potential targets for improving fertility are provided, enhancing potential to become pregnant either naturally or in a clinical setting.


Assuntos
Implantação do Embrião , Embrião de Mamíferos/metabolismo , Endométrio/metabolismo , Proteoma/metabolismo , Proteômica/métodos , Trofoblastos/metabolismo , Adulto , Linhagem Celular , Técnicas de Cocultura , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário , Endométrio/citologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Epitélio/metabolismo , Feminino , Fertilidade , Humanos , Gravidez , Trofoblastos/citologia
5.
Mol Hum Reprod ; 26(7): 498-509, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32449756

RESUMO

Current treatment options for uterine fibroids are limited to hormonal manipulation or surgical intervention. We aimed to develop an in vitro model to mirror collagen deposition and extracellular matrix (ECM) formation, the principal features of uterine fibroids, to enable testing of novel therapeutics. Macromolecular crowding with Ficoll 400 and Ficoll 70 in cultures of human uterine myometrial smooth muscle cells containing ascorbic acid, provided the basis for this model. These culture conditions mimic the 'crowded' nature of the in vivo extracellular environment by incorporating neutral, space-filling macromolecules into conventional cell cultures. This method of culture facilitates appropriate ECM deposition, thus closely representing the in vivo fibrotic phenotype of uterine fibroids. Macromolecular crowding in Ficoll cultures containing ascorbic acid reduced myometrial smooth muscle cell proliferation and promoted collagen production. Under these conditions, collagen was processed for extracellular deposition as demonstrated by C-propeptide cleavage from secreted procollagen. The fibrosis marker activin was increased relative to its natural inhibitor, follistatin, in crowded culture conditions while addition of exogenous follistatin reduced collagen (Col1A1) gene expression. This in vitro model represents a promising development for the testing of therapeutic interventions for uterine fibroids. However, it does not recapitulate the full in vivo pathology which can include specific genetic and epigenetic alterations that have not been identified in the myometrial smooth muscle (hTERT-HM) cell line. Following screening of potential therapeutics using the model, the most promising compounds will require further assessment in the context of individual subjects including those with genetic changes implicated in fibroid pathogenesis.


Assuntos
Matriz Extracelular/metabolismo , Leiomioma/metabolismo , Miócitos de Músculo Liso/metabolismo , Miométrio/metabolismo , Neoplasias Uterinas/metabolismo , Colágeno/metabolismo , Feminino , Humanos
6.
Hum Reprod ; 35(6): 1363-1376, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32488243

RESUMO

STUDY QUESTION: Does NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3) inflammasome activation within decidualized endometrial stromal cells accompany menstruation and is this reflected systemically? SUMMARY ANSWER: Components of the NLRP3 inflammasome immunolocalize to decidualized endometrial stromal cells immediately prior to menstruation, and are activated in an in vitro model of menstruation, as evidenced by downstream interleukin (IL)-1beta and IL-18 release, this being reflected systemically in vivo. WHAT IS KNOWN ALREADY: Menstruation is a highly inflammatory event associated with activation of NFκB (nuclear factor kappa-light-chain-enhancer of activated B cells), local release of chemokines and cytokines and inflammatory leukocyte influx. Systemically, chemokines and cytokines fluctuate across the menstrual cycle. STUDY DESIGN, SIZE, DURATION: This study examined the NLRP3 inflammasome and activation of downstream IL-1beta and IL-18 in endometrial tissues from women of known fertility (≥1 previous parous pregnancy) across the menstrual cycle (n ≥ 8 per cycle phase), serum from women during the proliferative, secretory and menstrual phases (≥9 per cycle phase) of the cycle and menstrual fluid collected on Day 2 of menses (n = 18). Endometrial stromal cells isolated from endometrial tissue biopsies (n = 10 in total) were used for an in vitro model of pre-menstrual hormone withdrawal. PARTICIPANTS/MATERIALS, SETTING, METHODS: Expression and localization of components of the NLRP3 inflammasome (NLRP3 & apoptosis-associated speck-caspase recruit domain [ASC]) in endometrial tissues was performed by immunohistochemistry. Unbiased digital quantification of immunohistochemical staining allowed determination of different patterns of expression across the menstrual cycle. Serum from women across the menstrual cycle was examined for IL-1beta and IL-18 concentrations by ELISA. An in vitro model of hormone withdrawal from estrogen/progestin decidualized endometrial stromal cells was used to more carefully examine activation of the NLRP3 inflammasome. Endometrial stromal cells isolated from endometrial tissue biopsies (n = 10) were treated with estrogen/medroxyprogesterone acetate for 12 days to induce decidualization (assessed by release of prolactin) followed by withdrawal of steroid hormone support. Activation of NLRP3, & ASC in these cells was examined on Days 0-3 after hormone withdrawal by Western immunoblotting. Release of IL-1beta and IL-18 examined during decidualization and across the same time course of hormone withdrawal by ELISA. Specific involvement of NLRP3 inflammasome activation in IL-1beta and IL-18 release after hormone withdrawal was investigated via application of the NLRP3 inflammasome inhibitor MCC950 at the time of hormone withdrawal. MAIN RESULTS AND THE ROLE OF CHANCE: Critical components of the NLRP3 inflammasome (NLRP3, ASC) were increased in menstrual phase endometrial tissues versus early secretory phase tissues (P < 0.05, n/s, respectively). NLRP3 and ASC were also elevated in the proliferative versus secretory phase of the cycle (P < 0.01, n/s, respectively) with ASC also significantly increased in the late-secretory versus early-secretory phase (P < 0.05). The pattern of activation was reflected in systemic levels of the inflammasome mediators, with IL-1beta and IL-18 elevated in peripheral blood serum during menstruation (Day 2 of menses) versus secretory phase (P = 0.026, P = 0.0042, respectively) and significantly elevated in menstrual fluid (Day 2 of menses) versus systemic levels across all cycle phases, suggesting that local inflammasome activation within the endometrium during menses is reflected by systemic inflammation. NLRP3 and ASC localized to decidualized cells adjacent to the spiral arterioles in the late secretory phase of the menstrual cycle, where the menstrual cascade is thought to be initiated, and to endometrial leukocytes during the menstrual phase. NLRP3 also localized to glandular epithelial cells during the late-secretory/menstrual phases. Localization of both NLRP3 and ASC switched from predominant epithelial localization during the early-secretory phase to stromal localization during the late-secretory/menstrual phase. Using an in vitro model of hormone withdrawal from decidualized human endometrial stromal cells, we demonstrated progressive activation of NLRP3 and ASC after hormone withdrawal increasing from Day 0 of withdrawal/Day 12 of decidualization to Day 3 of withdrawal. Downstream release of IL-1beta and IL-18 from decidualized stromal cells after hormone withdrawal followed the same pattern with the role of NLRP3 inflammasome activation confirmed via the inhibition of IL-1beta and IL-18 release upon application of MCC950. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: This study uses descriptive and semi-quantitative measures of NLRP3 inflammasome activation within endometrial tissues. Further, the in vitro model of pre-menstrual hormone withdrawal may not accurately recapitulate the in vivo environment as only one cell type is present and medroxyprogesterone acetate replaced natural progesterone due to its longer stability. WIDER IMPLICATIONS OF THE FINDINGS: We provide novel evidence that the NLRP3 inflammasome is activated within decidualized endometrial stromal cells immediately prior to menses and that local activation of the inflammasome within the endometrium appears to be reflected systemically in by activation of downstream IL-1beta and IL-18. Given the prevalence of menstrual disorders associated with inflammation including dysmenorrhoea and aspects of pre-menstrual syndrome, the inflammasome could be a novel target for ameliorating such burdens. STUDY FUNDING/COMPETING INTEREST(S): The authors have no competing interests. J.E. was supported by a Fielding Foundation fellowship, NHMRC project grants (#1139489 and #1141946) and The Hudson Institute of Medical Research. L.A.S. was supported by The Hudson Institute of Medical Research and J.H. by an Australian Government Research Training Program Scholarship. We acknowledge the Victorian Government's Operating Infrastructure funding to the Hudson Institute. TRIAL REGISTRATION NUMBER: N/A.


Assuntos
Inflamassomos , Menstruação , Austrália , Endométrio , Feminino , Humanos , Ciclo Menstrual , Gravidez
7.
FASEB J ; 33(1): 584-605, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30036086

RESUMO

Repair after damage is essential for tissue homeostasis. Postmenstrual endometrial repair is a cyclical manifestation of rapid, scar-free, tissue repair taking ∼3-5 d. Skin repair after wounding is slower (∼2 wk). In the case of chronic wounds, it takes months to years to restore integrity. Herein, the unique "rapid-repair" endometrial environment is translated to the "slower repair" skin environment. Menstrual fluid (MF), the milieu of postmenstrual endometrial repair, facilitates healing of endometrial and keratinocyte "wounds" in vitro, promoting cellular adhesion and migration, stimulates keratinocyte migration in an ex vivo human skin reconstruct model, and promotes re-epithelialization in an in vivo porcine wound model. Proteomic analysis of MF identified a large number of proteins: migration inhibitory factor, neutrophil gelatinase-associated lipocalin, follistatin like-1, chemokine ligand-20, and secretory leukocyte protease inhibitor were selected for further investigation. Functionally, they promote repair of endometrial and keratinocyte wounds by promoting migration. Translation of these and other MF factors into a migration-inducing treatment paradigm could provide novel treatments for tissue repair.-Evans, J., Infusini, G., McGovern, J., Cuttle, L., Webb, A., Nebl, T., Milla, L., Kimble, R., Kempf, M., Andrews, C. J., Leavesley, D., Salamonsen, L. A. Menstrual fluid factors facilitate tissue repair: identification and functional action in endometrial and skin repair.


Assuntos
Endométrio/citologia , Queratinócitos/citologia , Menstruação/metabolismo , Proteoma/metabolismo , Pele/citologia , Cicatrização , Animais , Adesão Celular , Movimento Celular , Proliferação de Células , Endométrio/metabolismo , Feminino , Humanos , Queratinócitos/metabolismo , Proteômica , Pele/metabolismo , Suínos
8.
Reprod Biomed Online ; 41(5): 757-766, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32972872

RESUMO

RESEARCH QUESTION: Proinflammatory advanced glycation end products (AGE), highly elevated within the uterine cavity of obese women, compromise endometrial function. Do AGE also impact preimplantation embryo development and function? DESIGN: Mouse embryos were cultured in AGE equimolar to uterine fluid concentrations in lean (1-2 µmol/l) or obese (4-8 µmol/l) women. Differential nuclear staining identified cell allocation to inner cell mass (ICM) and trophectoderm (TE) (day 4 and 5 of culture). Cell apoptosis was examined by terminal deoxynucleotidyl transferase-mediated dUDP nick-end labelling assay (day 5). Day 4 embryos were placed on bovine serum albumin/fibronectin-coated plates and embryo outgrowth assessed 93 h later as a marker of implantation potential. AGE effects on cell lineage allocation were reassessed following pharmacological interventions: either 12.5 nmol/l AGE receptor (RAGE) antagonist; 0.1 nmol/l metformin; or combination of 10 µmol/l acetyl-l-carnitine, 10 µmol/l N-acetyl-l-cysteine, and 5 µmol/l alpha-lipoic acid. RESULTS: 8 µmol/l AGE reduced: hatching rates (day 5, P < 0.01); total cell number (days 4, 5, P < 0.01); TE cell number (day 5, P < 0.01), and embryo outgrowth (P < 0.01). RAGE antagonism improved day 5 TE cell number. CONCLUSIONS: AGE equimolar with the obese uterine environment detrimentally impact preimplantation embryo development. In natural cycles, prolonged exposure to AGE may developmentally compromise embryos, whereas following assisted reproductive technology cycles, placement of a high-quality embryo into an adverse 'high AGE' environment may impede implantation success. The modest impact of short-term RAGE antagonism on improving embryo outcomes indicates preconception AGE reduction via pharmacological or dietary intervention may improve reproductive outcomes for overweight/obese women.


Assuntos
Desenvolvimento Embrionário/fisiologia , Produtos Finais de Glicação Avançada/metabolismo , Obesidade/metabolismo , Útero/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Benzamidas/farmacologia , Técnicas de Cultura Embrionária , Desenvolvimento Embrionário/efeitos dos fármacos , Feminino , Camundongos , Receptor para Produtos Finais de Glicação Avançada/antagonistas & inibidores , Útero/efeitos dos fármacos
9.
Reprod Fertil Dev ; 32(4): 363-372, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31839110

RESUMO

The menstruating spiny mouse is the first rodent identified to exhibit natural spontaneous decidualisation, cyclical endometrial shedding and regeneration. While the spiny mouse shares several primate-like characteristics in its reproductive biology, it has not been established whether pseudopregnancy can be induced or if its cycles can be synchronised as in non-human mammals. Here we describe attempts to induce pseudopregnancy and synchronisation of menstrual cycles (i.e. Whitten effect) in spiny mice. Virgin females (n=3-8 per group) underwent one of the following procedures to induce pseudopregnancy: daily vaginal lavage only (control), progesterone injection, mechanical stimulation of the cervix and sterile mating. A separate cohort was also exposed to male-soiled bedding to assess the Whitten effect. Pseudopregnancy was deemed successful if females presented with extended (>12 consecutive days) leukocytic vaginal cytology. No female from any method of induction met this criterion. In addition, the menstrual cycles of a group of six females could not be synchronised, nor immediate ovulation induced via exposure to male-soiled bedding. These responses indicate that the spiny mouse does not behave as a typical rodent. Like higher-order primates, the spiny mouse exhibits a relatively rare reproductive strategy, of failure to show pseudopregnancy or cyclical synchronisation. This is further endorsement of the use of this species as a versatile animal model for translational studies of menstruation and fertility.


Assuntos
Endométrio/fisiologia , Ciclo Menstrual , Ovulação , Pseudogravidez/fisiopatologia , Reprodução , Animais , Endométrio/efeitos dos fármacos , Feminino , Masculino , Ciclo Menstrual/efeitos dos fármacos , Murinae , Ovulação/efeitos dos fármacos , Periodicidade , Estimulação Física , Gravidez , Progesterona/administração & dosagem , Pseudogravidez/etiologia , Reprodução/efeitos dos fármacos , Especificidade da Espécie , Vasectomia
10.
J Assist Reprod Genet ; 37(1): 5-16, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31776756

RESUMO

OBJECTIVE: To establish a model of human implantation that responds to hormonal stimuli and can differentiate between endometrium from fertile women and those with idiopathic infertility. DESIGN: A trophoblast stem cell (trophectodermal) line (TSC; derived from human pre-implantation embryo) was used to form trophectodermal spheroids (TS). TS attachment to monolayers of endometrial epithelial cell lines or primary endometrial epithelial cells (pHEECs) was determined. SETTING: Independent Medical Research Institute with close clinical linkages INTERVENTIONS: Spheroid attachment and outgrowth was determined with added hormones (estradiol 17ß (E), E + medroxyprogesterone acetate (MPA) or E + MPA + human chorionic gonadotropin (hCG)). Spheroid attachment to E/MPA treated pHEEC prepared from fertile women or those with idiopathic infertility tested. MAIN OUTCOME MEASURE: Firmly attached spheroids counted after co-culture for 6 h. Outgrowth was determined by quantitation of area covered by spheroid after firm adhesion. RESULTS: Functional adhesion of TS to two endometrial epithelial cell lines, Ishikawa and ECC-1 cells, was hormonally responsive, with adhesion/outgrowth increased by E/MPA (ECC-1; p < 0.01, Ishikawa; p < 0.01) and E/MPA/hCG (ECC-1; p < 0.001, Ishikawa p < 0.01) versus E alone. The same pattern of hormone responsiveness was observed in pHEEC obtained from fertile women (E vs, E/MPA; p < 0.01, E vs. E/MPA/hCG; p < 0.001). TS adhered to 85% of pHEEC obtained from fertile women (11/13) and 11% of pHEEC obtained from women with unexplained infertility (2/18, p < 0.001). CONCLUSION: This new model of "embryo" implantation largely discriminates between endometrial epithelial cells obtained from fertile vs. infertile women based on adhesion; this holds potential as an in vitro "diagnostic" tool of endometrial infertility.


Assuntos
Adesão Celular , Embrião de Mamíferos/fisiologia , Endométrio/fisiologia , Estradiol/farmacologia , Fertilidade/fisiologia , Infertilidade Feminina/fisiopatologia , Trofoblastos/fisiologia , Técnicas de Cocultura , Implantação do Embrião , Embrião de Mamíferos/citologia , Embrião de Mamíferos/efeitos dos fármacos , Endométrio/citologia , Endométrio/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Estrogênios/farmacologia , Feminino , Fertilidade/efeitos dos fármacos , Humanos , Infertilidade Feminina/tratamento farmacológico , Esferoides Celulares/citologia , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/fisiologia , Trofoblastos/citologia , Trofoblastos/efeitos dos fármacos
11.
Proteomics ; 19(23): e1800423, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31531940

RESUMO

Embryo implantation into maternal endometrium is critical for initiation and establishment of pregnancy, requiring developmental synchrony between endometrium and blastocyst. However, factors regulating human endometrial-embryo cross talk and facilitate implantation remain largely unknown. Extracellular vesicles (EVs) are emerging as important mediators of this process. Here, a trophectoderm spheroid-based in vitro model mimicking the pre-implantation human embryo is used to recapitulate important functional aspects of blastocyst implantation. Functionally, human endometrial EVs, derived from hormonally treated cells synchronous with implantation, are readily internalized by trophectoderm cells, regulating adhesive and invasive capacity of human trophectoderm spheroids. To gain molecular insights into mechanisms underpinning endometrial EV-mediated enhancement of implantation, quantitative proteomics reveal critical alterations in trophectoderm cellular adhesion networks (cell adhesion molecule binding, cell-cell adhesion mediator activity, and cell adherens junctions) and metabolic and gene expression networks, and the soluble secretome from human trophectodermal spheroids. Importantly, transfer of endometrial EV cargo proteins to trophectoderm to mediate changes in trophectoderm function is demonstrated. This is highlighted by correlation among endometrial EVs, the trophectodermal proteome following EV uptake, and EV-mediated trophectodermal cellular proteome, important for implantation. This work provides an understanding into molecular mechanisms of endometrial EV-mediated regulation of human trophectoderm functions-fundamental in understanding human endometrium-embryo signaling during implantation.


Assuntos
Implantação do Embrião/fisiologia , Embrião de Mamíferos/metabolismo , Endométrio/metabolismo , Vesículas Extracelulares/metabolismo , Western Blotting , Adesão Celular/fisiologia , Microscopia Crioeletrônica , Células Epiteliais/metabolismo , Feminino , Humanos , Microscopia Eletrônica de Transmissão , Proteoma/metabolismo
12.
Hum Reprod ; 34(2): 308-322, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30561655

RESUMO

STUDY QUESTION: Does the newly discovered menstruating spiny mouse exhibit behavioural and metabolic changes in correlation with premenstrual phases of the menstrual cycle? SUMMARY ANSWER: This is the first report of cycle variability in the exploratory and interactive behaviour, and food consumption in menstruating spiny mice, and demonstrates that physiological changes are also dependent on within-subject variation. WHAT IS KNOWN ALREADY: Premenstrual syndrome (PMS) is a prominent cyclic disorder that affects millions of women worldwide. More than 70% of women endure symptoms of impending menstruation, such as bloating, abdominal cramping and nausea to some degree. Consequently, ~8% of women experience recurrent physical and emotional symptoms which are extreme enough to disrupt daily life and seek intervention. Due to a lack of an appropriate animal model, the mechanisms underlying PMS are poorly understood, and subsequently, effective treatments are limited. STUDY DESIGN, SIZE, DURATION: This study analyses the changes in behavioural responses to the investigator during vaginal lavage (n = 14), exploratory behaviour (n = 11) and metabolism (n = 20) across the menstrual cycle in the spiny mouse (Acomys cahirinus). PARTICIPANTS/MATERIALS, SETTING, METHODS: We performed vaginal lavages on virgin spiny mice (6-8 months of age) and subjected each cohort of females to repeated measures for vaginal lavage, exploratory behaviour and metabolism. Stages of the menstrual cycle were designated as early follicular, late follicular, early luteal, late luteal, early menstrual and late menstrual, with the late luteal and early menstrual phases considered as premenstrual phases and analysed using generalized estimating equations. For vaginal lavage, the behavioural responses to researcher handling were scored on an increasing scale of severity during the lavage process (e.g. restraint, frequency of vocalizations, total handling time). For exploratory behaviour, exploration, memory and sociability were assessed through subjection to Open Field (OF), Novel Object Recognition (NORT), Social Novelty (SN) and Elevated Plus Maze (EPM) tests. For metabolism, physiological changes were measured over a 24-h period in metabolic cages. Results are mean ± SD with statistical significance set to P < 0.05. MAIN RESULTS AND THE ROLE OF CHANCE: Qualitative behavioural assessment showed that compared to early follicular controls, during premenstrual phases, cycling females had significantly increased probability of: manifesting difficulties during restraint (4×, P < 0.01), vocalizing (8×, P < 0.01) and exhibiting isolation in the cage (40×, P = 0.041). We saw significant increases in handling time during the premenstrual phase in cycling females (76 ± 16 s) compared to controls (55 ± 7 s, P < 0.001). For exploratory behaviour, cycling females in their early menstrual phase travelled significantly less distance in the outer zone of the OF arena (13.3 ± 9.0 m) than females in their early luteal phase (22.3 ± 9.9 m, P = 0.038) and at significantly reduced velocities (40.2 ± 10.5 mm/s and 78.8 ± 31.0 mm/s, respectively, P = 0.006). These females also had fewer entries into the EPM open arms during the same phases (9.6 ± 6.1 and versus 20.0 ± 7.2, respectively, P = 0.030) and travelled less distance (3.2 ± 2.8 m versus 7.0 ± 5.5 m, respectively, P = 0.026). No differences were observed in NORT or SN across the cycle. In the metabolism studies, spiny mice demonstrated a significant increase in food consumption (percentage of body weight) during the early follicular and late luteal phases (3.9 ± 2.4% and 3.8 ± 2.1%, respectively) compared to the late follicular phase (2.3 ± 2.6%, P = 0.015). LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: This is an observational study to determine fundamental changes in behaviour and metabolism in a novel species, and as such, lacks commercially available laboratory reagents and protocols specific to the spiny mouse. WIDER IMPLICATIONS OF THE FINDINGS: The timing of these behavioural and physiological changes suggests that spiny mice exhibit symptoms analogous to PMS in higher order primates, thus providing a pre-clinical model for testing novel interventions to alleviate premenstrual symptoms and overcoming many limitations associated with this research area. STUDY FUNDING/COMPETING INTEREST(S): N.B. is supported by a Research Training Program stipend through Monash University. J.E. is supported by a Fellowship awarded by the Peter Fielding Foundation. The Hudson Institute of Medical Research is supported by the Victorian Government Operational Research Infrastructure Support. The authors declare no conflicts of interest.


Assuntos
Ingestão de Alimentos/fisiologia , Comportamento Exploratório/fisiologia , Ciclo Menstrual/fisiologia , Síndrome Pré-Menstrual/fisiopatologia , Animais , Técnicas de Observação do Comportamento , Comportamento Animal/fisiologia , Variação Biológica da População , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Síndrome Pré-Menstrual/diagnóstico
13.
J Assist Reprod Genet ; 36(5): 811-817, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30610663

RESUMO

The common spiny mouse (Acomys cahirinus) is the only known rodent to demonstrate a myriad of physiological processes unseen in their murid relatives. The most recently discovered of these uncharacteristic traits: spontaneous decidual transformation of the uterus in virgin females, preceding menstruation. Menstruation occurring without experimental intervention in rodents has not been documented elsewhere to date, and natural menstruation is indeed rare in the animal kingdom outside of higher order primates. This review briefly summarises the current knowledge of spiny mouse biology and taxonomy, and explores their endocrinology which may aid in our understanding of the evolution of menstruation in this species. We propose that DHEA, synthesised by the spiny mouse (but not other rodents), humans and other menstruating primates, is integral in spontaneous decidualisation and therefore menstruation. We discuss both physiological and behavioural attributes across the menstrual cycle in the spiny mouse analogous to those observed in other menstruating species, including premenstrual syndrome. We further encourage the use of the spiny mouse as a small animal model of menstruation and female reproductive biology.


Assuntos
Decídua/fisiologia , Menstruação , Murinae/fisiologia , Animais , Decídua/citologia , Feminino , Haplorrinos , Humanos , Camundongos
14.
Biol Reprod ; 98(6): 752-764, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29546322

RESUMO

The regenerative, proliferative phase of a woman's menstrual cycle is a critical period which lays the foundation for the subsequent, receptive secretory phase. Although endometrial glands and their secretions are essential for embryo implantation and survival, the proliferative phase, when these glands form, has been rarely examined. We hypothesized that alterations in the secreted proteome of the endometrium of idiopathic infertile women would reflect a disturbance in proliferative phase endometrial regeneration. Our aim was to compare the proteomic profile of proliferative phase uterine fluid from fertile (n = 9) and idiopathic infertile (n = 10) women. Proteins with ≥2-fold change (P < 0.05) were considered significantly altered between fertile and infertile groups. Immunohistochemistry examined the endometrial localization of identified proteins. Western immunoblotting defined the forms of extracellular matrix protein 1 (ECM1) in uterine lavage fluid. Proteomic analysis identified four proteins significantly downregulated in infertile women compared to fertile women, including secreted frizzled-related protein 4 (SFRP4), CD44, and ECM1: two proteins were upregulated. Seven proteins were unique to the fertile group and six (including isoaspartyl peptidase/L-asparaginase [ASRGL1]) were unique to the infertile group. Identified proteins were classified into biological processes of tissue regeneration and regulatory processes. ASRGL1, SFRP4, and ECM1 localized to glandular epithelium and stroma, cluster of differentiation 44 (CD44) to stroma and immune cells. ECM1 was present in two main molecular weight forms in uterine fluid. Our results indicate a disturbance in endometrial development during the proliferative phase among infertile women, providing insights into human endometrial development and potential therapeutic targets for infertility.


Assuntos
Líquidos Corporais/metabolismo , Endométrio/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Fase Folicular/metabolismo , Receptores de Hialuronatos/metabolismo , Infertilidade Feminina/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Adulto , Feminino , Regulação da Expressão Gênica , Humanos , Proteômica
15.
Hum Reprod ; 33(4): 654-665, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29471449

RESUMO

STUDY QUESTION: Do obese levels of advanced glycation end products (AGEs) within the uterine cavity detrimentally alter tissue function in embryo implantation and placental development? SUMMARY ANSWER: Obese levels of AGEs activate inflammatory signaling (p65 NFκB) within endometrial epithelial cells and alter their function, cause endoplasmic reticulum (ER) stress in endometrial stromal cells and impair decidualization, compromise implantation of blastocyst mimics and inhibit trophoblast invasion. WHAT IS KNOWN ALREADY: Obese women experience a higher incidence of infertility, recurrent miscarriage and pregnancy complications compared with lean women. Oocyte donation cycles suggest a detrimental uterine environment plays a role in these outcomes. STUDY DESIGN, SIZE, DURATION: Uterine lavage and tissues from lean (BMI 19.5-24.9, n = 17) and obese (BMI > 30, n = 16) women examined. Cell culture experiments utilizing human endometrial epithelial, trophectoderm and trophoblast cell lines and primary human stromal cells used to examine the functional impact of obese levels of AGEs. PARTICIPANTS/MATERIALS, SETTING, METHODS: Levels of AGEs examined within uterine lavage assessed by ELISA to determine differences between lean and obese women. Expression and localization of AGEs, receptor for AGEs (RAGE) and NFκB within endometrial tissues obtained from lean and obese women determined by immunohistochemistry. Endometrial epithelial cells (ECC-1), primary human stromal cells and trophoblast cells (HTR8-SVneo) treated with lean (2000 nmol/mol lysine) or obese (8000 nmol/mol lysine) uterine levels of AGEs and p65 NFκB (western immunoblot), real-time adhesion, proliferation migration and invasion (xCelligence real-time cell function analysis), decidualization (cell morphology and prolactin release), ER stress (western immunoblot for p-PERK) determined. Co-cultures of endometrial epithelial cells and blastocyst mimics (trophectoderm spheroids) similarly treated with lean or obese uterine levels of AGEs to determine their impact on embryo implantation. MAIN RESULTS AND THE ROLE OF CHANCE: AGEs were significantly elevated (P = 0.004) within the obese (6503.59 µmol/mol lysine) versus lean (2165.88 µmol/mol lysine) uterine cavity (uterine lavage) with increased immunostaining for AGEs, RAGE and NFkB within obese endometrial tissues during the proliferative phase of the menstrual cycle. Obese uterine levels of AGEs inhibited adhesion and proliferation of endometrial epithelial (ECC-1) cells compared to treatment with lean uterine levels of AGEs. Obese uterine AGE levels impacted primary human endometrial stromal cell decidualization and activated ER stress within these cells. Obese uterine levels of AGEs also inhibited trophectodermal spheroid adhesion to hormonally primed endometrial epithelial cells and trophoblast cell line HTR8/SV-neo invasion. LARGE SCALE DATA: N/A. LIMITATIONS REASONS FOR CAUTION: Mechanistic studies are performed in vitro and may not completely recapitulate cell function in vivo. WIDER IMPLICATIONS OF THE FINDINGS: These data corroborate clinical data suggesting the presence of an altered uterine environment in obese women and demonstrate that elevated uterine levels of AGEs within these women may detrimentally impact endometrial function, embryo implantation and placental development. Uterine AGE assessment in infertility work up may prove useful in determining underlying causes of infertility. AGEs can be targeted pharmacologically and such treatments may prove effective in improving reproductive complications experience by obese women. STUDY FUNDING/COMPETING INTEREST(S): Supported by NHMRC Fellowship (#1002028 to L.A.S.), and the Victorian Government's Operational Infrastructure Support Program. MTC is supported by a JDRF Australia Clinical Research Network Career Development Award. The authors have declared that no conflict of interest exists.


Assuntos
Implantação do Embrião/fisiologia , Endométrio/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Obesidade/metabolismo , Útero/metabolismo , Adulto , Células Epiteliais/metabolismo , Feminino , Humanos , Placentação/fisiologia , Gravidez , Células Estromais/metabolismo , Trofoblastos/metabolismo
16.
Hum Reprod ; 33(9): 1715-1726, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-30032205

RESUMO

STUDY QUESTION: Is the newly discovered menstruating rodent, the spiny mouse, a valid model for studying endometrial morphology and menstruation? SUMMARY ANSWER: Our study is the first to demonstrate a primate-like pattern of natural menstruation in a rodent, with decidualization, spiral arteriole remodeling and piece-meal endometrial shedding. WHAT IS KNOWN ALREADY: The spiny mouse has a naturally occurring menstrual cycle. This unique feature has the potential to reduce the heavy reliance on primates and provide a more appropriate small animal model for menstrual physiology research. STUDY DESIGN, SIZE, DURATION: This study compares morphological changes in the endometrium during early, mid and late menstruation of the spiny mouse (n = 39), human (n = 9) and the induced mouse model of menstruation (n = 17). PARTICIPANTS/MATERIALS, SETTING, METHODS: We assessed tissue morphology with hematoxylin and eosin and erythrocyte patterns with Mallory's trichrome. We conducted staining for neutrophil gelatinase associated lipocalin (NGAL), cytokeratin and interleukin-11 (IL-11) in all species. We used double immunofluorescence staining for vascular endothelial growth factor and alpha-smooth muscle actin to detect vasculature remodeling and western immunoblot to detect interleukin-8 (IL-8) and macrophage migration inhibitory factor (MIF) in the menstrual fluid of spiny mice. MAIN RESULTS AND THE ROLE OF CHANCE: Menstruation occurs in the spiny mouse over a 72-h period, with heaviest menstrual breakdown occurring 24 h after initial observation of red blood cells in the vaginal cytology. During menstruation, the endometrium of the spiny mouse appeared to resemble human menstrual shedding with focal epithelial breakdown observed in conjunction with lysis of underlying stroma and detection of IL-8 and MIF in menstrual fluid. The mouse exhibits extensive decidualization prior to induced menses, with transformation of the entire uterine horn and cytokeratin expression absent until initiation of repair. Decidualization occurred spontaneously and was less marked in the spiny mouse, where epithelial integrity remained intact. In all species, the decidua was positive for IL-11 secretion and neutrophil recruitment was similar in each. Spiral arteriole formation was confirmed in the spiny mouse. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: This is a descriptive study comparing primarily morphological traits between the spiny mouse, the mouse and the human. Reagents specific to the spiny mouse were limited and resources for global use of this novel species are few. WIDER IMPLICATIONS OF THE FINDINGS: Our work supports the spiny mouse as a viable model, sharing many attributes of physiological menstruation with humans. The strength of a natural as opposed to an artificial model is validated through the striking similarities observed between the spiny mouse and human in uterine breakdown. The spiny mouse may be highly useful in large-scale investigations of menstruation and menstrual disorders. STUDY FUNDING/COMPETING INTEREST(S): N.B. and S.R. are each recipients of a Research Training Program scholarship supported by Monash University. This work was supported by the Victorian Government Operational Infrastructure and laboratory funds to H.D. The authors declare no competing interests.


Assuntos
Decídua/metabolismo , Menstruação/metabolismo , Murinae , Animais , Western Blotting , Decídua/citologia , Feminino , Humanos , Fatores Inibidores da Migração de Macrófagos/sangue , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Fator A de Crescimento do Endotélio Vascular/sangue
17.
Cytokine ; 111: 222-229, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30195213

RESUMO

The endometrium lines a women's uterus becoming receptive, and allowing embryo implantation to occur, for just a few days during the post-ovulatory mid-secretory phase of each menstrual cycle. We investigated whether concentrations of proposed receptivity biomarkers (VEGF, IL8, FGF2, CSF3 sFlt-1, sGP130 and PlGF) secreted by the endometrium into the uterine cavity and forming the microenvironment for embryo implantation is altered among a population of age-matched women with unexplained (idiopathic) infertility compared to fertile women during the receptive mid-secretory phase (n = 16 fertile, 18 infertile) and the prior pre-receptive early secretory phase (n = 19 fertile, 18 infertile) of their cycle. In the mid-secretory cohort significantly elevated concentrations of five biomarkers; PlGF (p = 0.001), IL8 (p = 0.004), sGP130 (p = 0.009), sFlt-1 (p = 0.021), and CSF3 (p = 0.029) was present in uterine fluid of infertile women during the mid-secretory phase, but only CSF3 was significantly elevated in the pre-receptive early secretory phase (p = 0.006). In vitro studies of glycosylated and non-glycosylated forms of CSF3 at representative fertile (20 ng/mL) and infertile (70 ng/mL) effects on endometrium and embryo behaviour were performed. Non-glycosylated CSF3 at fertile concentrations significantly (p < 0.001) elevated endometrial epithelial cell proliferation however chronic treatment or elevated (infertile) concentrations of CSF3 in glycosylated form abrogated the positive effects. Both forms of CSF3 increased trophoblast cell invasion (p < 0.001) regardless of concentration. Mouse embryo outgrowth was significantly (p < 0.01) increased at fertile but not at infertile concentrations. The study confirmed potential utility of five biomarkers of endometrial receptivity for future application in the mid-secretory phase while highlighting CSF3 is elevated in the earlier pre-receptive phase. Our data provides evidence that CSF3 acts on both human endometrium and embryo in a manner that is concentration and glycosylation dependent.


Assuntos
Biomarcadores/metabolismo , Endométrio/metabolismo , Útero/metabolismo , Animais , Linhagem Celular , Microambiente Celular/fisiologia , Estudos de Coortes , Implantação do Embrião/fisiologia , Feminino , Fertilidade/fisiologia , Humanos , Infertilidade Feminina/metabolismo , Ciclo Menstrual/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
18.
Biol Reprod ; 92(4): 99, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25715794

RESUMO

The endometrium (the mucosal lining of the uterus) is a dynamic tissue that undergoes extensive remodeling, secretory transformation in preparation for implantation of an embryo, inflammatory and proteolytic activity during menstruation, and rapid postmenstrual repair. A plethora of local factors influence these processes. Recently, a cysteine-rich protein, CRISP3, a clade of the CRISP, antigen 5, pathogenesis-related (CAP) protein superfamily, has been implicated in uterine function. The localization, regulation, and potential function of CRISP3 in both the human and mouse endometrium is described. CRISP3 localizes to the luminal and glandular epithelium of the endometrium within both species, with increased immunoreactivity during the proliferative phase of the human cycle. CRISP3 also localizes to neutrophils, particularly within the premenstrual human endometrium and during the postbreakdown repair phase of a mouse model of endometrial breakdown and repair. Endometrial CRISP3 is produced by primary human endometrial epithelial cells and secreted in vivo to accumulate in the uterine cavity. Secreted CRISP3 is more abundant in uterine lavage fluid during the proliferative phase of the menstrual cycle. Human endometrial epithelial CRISP3 is present in both a glycosylated and a nonglycosylated form in vitro and in vivo. Treatment of endometrial epithelial cells in vitro with recombinant CRISP3 enhances both adhesion and proliferation. These data suggest roles for epithelial and neutrophil-derived CRISP3 in postmenstrual endometrial repair and regeneration.


Assuntos
Adesão Celular/fisiologia , Endométrio/fisiologia , Células Epiteliais/fisiologia , Ciclo Estral/fisiologia , Ciclo Menstrual/fisiologia , Proteínas e Peptídeos Salivares/biossíntese , Proteínas de Plasma Seminal/biossíntese , Adulto , Animais , Proliferação de Células , Endométrio/citologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/metabolismo , Gravidez , Cultura Primária de Células , Proteínas e Peptídeos Salivares/genética , Proteínas de Plasma Seminal/genética
19.
Biol Reprod ; 90(1): 14, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24227758

RESUMO

Menstruation is a complex process dependent on premenstrual release of inflammatory mediators and proteolytic enzymes from endometrial cells. Endometrial leukocytes are traditionally considered to be the major source of the inflammatory factors. However, evidence is emerging to suggest a role for decidualized endometrial stromal cells in the premenstrual inflammatory cascade. We sought to determine if withdrawal of hormone support (estrogen and progesterone) from decidualized endometrial stromal cells, in a model mimicking the precise timing leading to menstruation, activated inflammatory signaling pathways and downstream release of inflammatory mediators. Human endometrial stromal cells decidualized gradually over 12 days of estradiol and progestin treatment as evidenced by an increase in prolactin secretion. Withdrawal of hormone support from decidualized stromal cells resulted in a decrease in cytoplasmic IkappaB and a progressive increase in nuclear accumulation of NF-kappaB, as demonstrated by Western immunoblot and immunocytochemical analyses. Concomitant with nuclear translocation of NF-kappaB, hormone withdrawal led to production of a host of inflammatory mediators by the decidualized stromal cells, including IFN-alpha, IL-6, CCL11, GM-CSF, CCL2, IL1-RA, CXCL10, CXCL8, IL-12, IL-15, VEGF, and CCL5. Elevation of inflammatory mediators was not observed, however, upon hormone withdrawal in cells treated with the NF-kappaB inhibitor BAY 11-7085. Decidualized stromal cells are likely highly sensitive sensors of changing hormone levels. This provides a mechanism by which decidualized stromal cells may recruit inflammatory leukocytes into the premenstrual endometrium and contribute to the intense inflammation underlying this unique physiological process.


Assuntos
Decídua/fisiologia , Endométrio/fisiologia , Estradiol/farmacologia , Inflamação/metabolismo , Ciclo Menstrual/efeitos dos fármacos , Progesterona/farmacologia , Células Estromais/fisiologia , Células Cultivadas , Quimiotaxia de Leucócito/efeitos dos fármacos , Quimiotaxia de Leucócito/fisiologia , Endométrio/citologia , Estradiol/deficiência , Estradiol/metabolismo , Feminino , Humanos , Ciclo Menstrual/imunologia , Ciclo Menstrual/metabolismo , NF-kappa B/antagonistas & inibidores , Nitrilas/farmacologia , Progesterona/deficiência , Progesterona/metabolismo , Sulfonas/farmacologia
20.
Mol Hum Reprod ; 20(8): 787-98, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24782449

RESUMO

Menstruation involves the shedding of the functional layer of the endometrium in the absence of pregnancy. At sites where tissue shedding is complete, re-epithelialization of the tissue is essential for repair and termination of bleeding. The complement of growth factors that mediate post-menstrual endometrial repair are yet to be completely elucidated. Galectins regulate many cell functions important for post-menstrual repair, such as cell adhesion and migration. Galectin-7 has a well characterized role in re-epithelialization and wound healing. We hypothesized that galectin-7 would be important in re-epithelialization during post-menstrual repair. We aimed to identify endometrial expression of galectin-7 in women undergoing normal endometrial repair and in women with amenorrhoea who do not experience endometrial breakdown and repair, and to determine whether galectin-7 enhances endometrial re-epithelialization in vitro. Galectin-7 immunolocalized to the endometrial luminal and glandular epithelium during the late secretory and menstrual phases, and to decidualized stroma in regions exhibiting tissue breakdown. Immunostaining intensity was significantly reduced in the endometrium of women with amenorrhoea compared with normally cycling woman. ELISA identified galectin-7 in menstrual fluid at significantly elevated levels compared with matched peripheral plasma. Exogenous galectin-7 (2.5 µg/ml) significantly enhanced endometrial epithelial wound repair in vitro; this was abrogated by inhibition of integrin binding. Galectin-7 elevated epithelial expression of extracellular matrix-related molecules likely involved in repair including ß-catenin, contactin and TGF-ß1. In conclusion, galectin-7 is produced by the premenstrual and menstrual endometrium, where it accumulates in menstrual fluid and likely acts as a paracrine factor to facilitate post-menstrual endometrial re-epithelialization.


Assuntos
Endométrio/metabolismo , Galectinas/metabolismo , Ciclo Menstrual/metabolismo , Menstruação/metabolismo , Células Cultivadas , Endométrio/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Feminino , Galectinas/genética , Galectinas/farmacologia , Humanos , Técnicas In Vitro , Ciclo Menstrual/efeitos dos fármacos , Gravidez , Fator de Crescimento Transformador beta1/metabolismo , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA