Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Pathol ; 190(1): 145-157, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31610174

RESUMEN

Repeated tissue injury and repair and fibrosis play a pivotal role in endometriosis. Fibrotic tissue consists of extracellular matrix proteins, regulated by transcriptional factors promoting cell proliferation and survival. Periostin is one of the putative key extracellular matrix proteins. This study aimed to determine whether transcription factor 21 (TCF21) is involved in the development of endometriosis as an upstream regulatory gene of periostin. Formalin-fixed, paraffin-embedded tissue samples [normal endometrium of women without endometriosis; eutopic endometrium of women with endometriosis; ovarian endometriosis (OE); and deep infiltrating endometriosis (DIE)] and respective cells were analyzed. Basal, transiently stimulated, and knocked down periostin and TCF21 concentrations in stromal cells of women with or without endometriosis were examined. Periostin and TCF21 expressions were undetected in normal endometrium of women without endometriosis, weakly positive in eutopic endometrium of women with endometriosis, moderately positive in OE, and strongly positive in DIE. Type 2 helper T-cell cytokines (IL-4, IL-13, and transforming growth factor-ß1) increased the mRNA expression of periostin and TCF21. These cytokines, periostin, and TCF21 colocalized in the stroma of OE and DIE. siRNA against human TCF21 gene suppressed periostin expression. Transfection of TCF21 plasmid vector into stromal cells of women without endometriosis, which originally expressed neither periostin nor TCF21, resulted in TCF21 and periostin expression. TCF21 and periostin are involved in the regulation of fibrosis in endometriosis. TCF21 may be a promising therapeutic target and biomarker in endometriosis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Moléculas de Adhesión Celular/metabolismo , Endometriosis/patología , Endometrio/patología , Fibrosis/patología , Células del Estroma/patología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Biomarcadores , Estudios de Casos y Controles , Moléculas de Adhesión Celular/genética , Proliferación Celular , Células Cultivadas , Citocinas , Endometriosis/genética , Endometriosis/metabolismo , Endometrio/metabolismo , Femenino , Fibrosis/genética , Fibrosis/metabolismo , Humanos , Células del Estroma/metabolismo
2.
Reproduction ; 162(4): 277-287, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34324431

RESUMEN

This study aimed to clarify the physiological mechanism regulating the growth of primordial follicles in mouse ovaries. In a previous study, we found that increasing the fetal bovine serum concentration in the culture medium promoted the growth of primordial follicles in cultured postnatal day 0 ovaries but not in cultured postnatal day 4 ovaries. Based on these results, we hypothesized that the regulatory system repressing the growth of primordial follicles is established in postnatal day 4 ovaries. To confirm this hypothesis, microarray analysis of postnatal day 0 and 4 ovaries was performed. The results revealed that the expression of mRNA of stefin A homologs increased in postnatal day 4 ovaries. Stefin A belonging to the type 1 cystatin superfamily is an inhibitor of cysteine cathepsins. Consistently, the inhibitor of cathepsins repressed the growth of primordial follicles in cultured postnatal day 0 ovaries. Furthermore, we found that 17ß-estradiol promoted the expression of mRNA of stefin A homologs in cultured ovaries and repressed the growth of primordial follicles. Our results demonstrate that 17ß-estradiol and cathepsins regulate the growth of primordial follicles in mouse ovaries.


Asunto(s)
Catepsinas , Ovario , Animales , Catepsinas/genética , Catepsinas/metabolismo , Estradiol/metabolismo , Estradiol/farmacología , Femenino , Ratones , Folículo Ovárico/metabolismo , Ovario/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
3.
Reproduction ; 158(1): 25-34, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30933929

RESUMEN

Folliculogenesis is a complex process, defined by the growth and development of follicles from the primordial population. Granulosa cells (GCs) play a vital role in every stage of follicular growth through proliferation, acquisition of gonadotropic responsiveness, steroidogenesis and production of autocrine/paracrine factors. A recently discovered hypothalamic neuropeptide phoenixin is involved in the regulation of the reproductive system. Phoenixin acts through its receptor, G protein-coupled receptor 173 (GPR173), to activate the cAMP/PKA pathway leading to the phosphorylation of CREB (pCREB). Here, we demonstrated the expression patterns of phoenixin and GPR173 in human ovary and explored its role in folliculogenesis. Phoenixin and GPR173 were both expressed in the human ovarian follicle, with increased expression in GCs as the follicle grows. Phoenixin treatment at 100 nM for 24 h induced the proliferation of human non-luteinized granulosa cell line, HGrC1 and significantly increased the expression levels of CYP19A1, FSHR, LHR and KITL, but decreased NPPC expression levels. These effects were suppressed by GPR173 siRNA. The expression level of CREB1, pCREB and estradiol (E2) production in the culture medium was significantly enhanced by phoenixin treatment in a concentration-dependent manner. Phoenixin also significantly increased the follicular area in a murine ovarian tissue culture model, leading to an increased number of ovulated oocytes with a higher level of maturation. Taken together, our data demonstrate that phoenixin is an intraovarian factor that promotes follicular growth through its receptor GPR173 by accelerating proliferation of GCs, inducing E2 production and increasing the expression of genes related to follicle development.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células de la Granulosa/citología , Neuropéptidos/farmacología , Folículo Ovárico/citología , Receptores Acoplados a Proteínas G/metabolismo , Adulto , Animales , Femenino , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Humanos , Ratones , Neuropéptidos/análisis , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/metabolismo , Receptores Acoplados a Proteínas G/genética , Adulto Joven
4.
J Assist Reprod Genet ; 36(6): 1185-1194, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31079267

RESUMEN

PURPOSE: Follicle-stimulating hormone receptor (FSHR) expression in granulosa cells is critical in enabling follicles to achieve accelerated growth. Although FSHR expression has been reported to be epigenetically regulated, the mechanism is unclear. Cooperation between oocytes and granulosa cells is also essential for normal follicular growth. Among oocyte-derived factors, bone morphogenetic protein 15 (BMP15) promotes follicular growth and is suggested to have epigenetic effects. We examined the role of BMP15 in the acquirement of FSHR in human granulosa cells. METHODS: Immortalized non-luteinized human granulosa (HGrC1) cells were stimulated with trichostatin A (TSA) or BMP15 to analyze FSHR expression, histone modifications, and USF1/2 binding at the FSHR promoter region. Histone acetyl transferase (HAT) activity and phosphorylation of Smad 1/5/8 and p38 MAPK were examined with or without BMP15, SB203580, and LDN193189. CYP19A1 expression and estradiol production were also studied. RESULTS: TSA and BMP15 induced FSHR mRNA expression in a dose-dependent manner and histone modifications were observed with increased binding of USF1/2. BMP15 increased FSHR protein expression, which was suppressed by LDN193189. BMP15 increased phosphorylation of Smad 1/5/8 and significantly increased HAT activity, which was inhibited by LDN193189, but not by SB203580. BMP15 increased phosphorylation of p38 MAPK and USF1. LDN193189 suppressed BMP15-induced phosphorylation of both p38 MAPK and USF1, whereas SB203580 suppressed the phosphorylation of USF1. BMP15 increased CYP19A1 mRNA expression and estradiol production. CONCLUSION: BMP15 induced FSHR expression in human granulosa cells through Smad and non-Smad pathways. This mechanism of FSHR induction by BMP15 may be utilized for controlling follicular growth.


Asunto(s)
Proteína Morfogenética Ósea 15/farmacología , Células de la Granulosa/metabolismo , Oocitos/crecimiento & desarrollo , Receptores de HFE/genética , Aromatasa/genética , Estradiol/genética , Femenino , Hormona Folículo Estimulante/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células de la Granulosa/patología , Humanos , Ácidos Hidroxámicos/farmacología , Oocitos/metabolismo , Folículo Ovárico/crecimiento & desarrollo , Folículo Ovárico/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas Smad/genética
5.
Reprod Med Biol ; 18(2): 151-160, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30996678

RESUMEN

BACKGROUND: Polycystic ovary syndrome (PCOS) is a common endocrine disorder among women of reproductive age and a major cause of infertility; however, the pathophysiology of this syndrome is not fully understood. This can be addressed using appropriate animal models of PCOS. In this review, we describe rodent models of hormone-induced PCOS that focus on the perturbation of the hypothalamic-pituitary-ovary (HPO) axis and abnormalities in neuropeptide levels. METHODS: Comparison of rodent models of hormone-induced PCOS. MAIN FINDINGS: The main method used to generate rodent models of PCOS was subcutaneous injection or implantation of androgens, estrogens, antiprogestin, or aromatase inhibitor. Androgens were administered to animals pre- or postnatally. Alterations in the levels of kisspeptin and related molecules have been reported in these models. CONCLUSION: The most appropriate model for the research objective and hypothesis should be established. Dysregulation of the HPO axis followed by elevated serum luteinizing hormone levels, hyperandrogenism, and metabolic disturbance contribute to the complex etiology of PCOS. These phenotypes of the human disease are recapitulated in hormone-induced PCOS models. Thus, evidence from animal models can help to clarify the pathophysiology of PCOS.

6.
Reprod Biol Endocrinol ; 16(1): 106, 2018 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-30368246

RESUMEN

BACKGROUND: Given the seriousness of chemotherapy-induced ovarian injury in female cancer patients, the preservation of fertility, including through the use of cryopreservation technology and pharmaceuticals, requires investigation. Previous studies have shown that damage to the ovaries is related to oxidative stress caused by anticancer drugs. Therefore, superoxide dismutase (SOD) may represent a key factor in the pharmacological protection of the ovaries. The aim of our study was to identify the effects of mangafodipir, a manganese chelate and SOD-mimetic, on suppression of apoptosis in granulosa cells and primordial follicle activation induced by anticancer drugs. METHODS: Cell viability assays using methyltrichlorosilane solutions and immunoblotting for cleaved caspase-3 were performed in in vitro experiments with the simultaneous addition of mangafodipir to human non-luteinized granulosa cell line (HGrC) cultures treated with hydrogen peroxide (H2O2), cisplatin, or paclitaxel. Count and morphological analyses of follicles at each developing stage in the ovaries and immunohistochemistry for cleaved caspase-3, Ki67 and 4-hydroxynonenal, a marker for oxidative stress, were also performed using mangafodipir-injected 6-week-old female ICR mice treated with cisplatin or paclitaxel. Further, mangafodipir was injected into 6-week-old female BALB/c mice inoculated with ES-2 to analyze whether mangafodipir inhibits the anti-tumor effects of cisplatin or paclitaxel treatment. RESULTS: Mangafodipir attenuated apoptosis induced by H2O2 and anticancer drugs in vitro. Mangafodipir also decreased the expression of 4-hydroxynonenal and reduced cisplatin- and paclitaxel-induced apoptosis in granulosa cells in vivo. In addition, mangafodipir inhibited the loss of primordial follicles. Tumor xenograft studies in mice showed that mangafodipir did not affect anticancer drug antitumor effects. CONCLUSIONS: Oxidative stress might be one of the mechanisms of cisplatin- and paclitaxel-induced the loss of primordial follicles. Mangafodipir can reduce cisplatin- and paclitaxel-induced apoptosis in granulosa cells and primordial follicle activation partially via its SOD activity. At the same time, mangafodipir might have other potential mechanisms to inhibit the activation of primordial follicles. Further, mangafodipir attenuated the ovarian damage caused by cisplatin and paclitaxel without affecting their antitumor activities. Mangafodipir, therefore, though its efficacy might be limited, may be a new option for the preservation of fertility during anticancer treatment.


Asunto(s)
Antineoplásicos/farmacología , Ácido Edético/análogos & derivados , Células de la Granulosa/efectos de los fármacos , Ovario/efectos de los fármacos , Fosfato de Piridoxal/análogos & derivados , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular , Línea Celular Tumoral , Cisplatino/farmacología , Medios de Contraste/farmacología , Ácido Edético/farmacología , Femenino , Células de la Granulosa/citología , Células de la Granulosa/metabolismo , Humanos , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Ratones Desnudos , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/metabolismo , Ovario/metabolismo , Estrés Oxidativo/efectos de los fármacos , Paclitaxel/farmacología , Sustancias Protectoras/farmacología , Fosfato de Piridoxal/farmacología
7.
Horm Metab Res ; 50(7): 537-542, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29991084

RESUMEN

Patients with primary ovarian insufficiency (POI) have a high prevalence of thyroid autoimmune disorders. However, the extent of the contribution of thyroid autoantibodies or elevated thyroid-stimulating hormone (TSH) levels to decreased ovarian reserve is unclear. Therefore, we evaluated the serum levels of anti-Müllerian hormone (AMH) and thyroid autoantibodies [antithyroperoxidase antibody (TPOAb), and antithyroglobulin antibody (TgAb)] in euthyroid infertile women. One hundred and fifty-three women with normal menstrual cycles were recruited for this retrospective study. Serum levels of AMH were compared between patients with positive and negative thyroid autoantibodies. The correlation between serum levels of AMH and each thyroid autoantibody was also evaluated. Participants were observed to be either TPOAb or TgAb positive (n=27), only TPOAb positive (n=8), only TgAb positive (n=7), TPOAb and TgAb positive (double positive; n=12), and TPOAb and TgAb negative (double negative; n=126). No significant differences were found in serum AMH levels between the TPOAb- or TgAb-positive women and the antibody-double negative women. Serum AMH levels did not show a significant correlation with the concentration of TgAb or TPOAb. On the other hand, serum AMH levels negatively correlated with TSH levels in patients who were either positive for TPOAb or TgAb. Thyroid autoantibodies are not likely to influence ovarian reserve in euthyroid women whose TSH levels fall within the normal range although elevated TSH levels may be involved in the decline of serum AMH levels.


Asunto(s)
Autoanticuerpos/sangre , Infertilidad Femenina/fisiopatología , Reserva Ovárica/fisiología , Glándula Tiroides/fisiopatología , Adulto , Estudios Transversales , Femenino , Humanos , Ciclo Menstrual , Estudios Retrospectivos
8.
J Obstet Gynaecol Res ; 44(6): 998-1006, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29517134

RESUMEN

It has been more than 15 years since the measurement of serum anti-Müllerian hormone (AMH) first allowed the quantitative assessment of ovarian reserve. Meanwhile, the clinical implication of serum AMH has been expanding. The measurement of serum AMH has been applied in various clinical fields, including assisted reproduction, menopause, reproductive disorders and assessment of ovarian damage/toxicity. Well-known findings about the usefulness of serum AMH revealed by numerous studies executed in the early era include decline with aging, a good correlation with oocyte yield in assisted reproduction, upregulation in polycystic ovarian syndrome and a decrease on ovarian surgery and toxic treatment. More intensive research, including a meta-analysis, cutting-edge clinical trial and advances in AMH assays, has yielded newer findings and firmer clinical interpretations in serum AMH in the past few years. Variations in the AMH decline trajectory in the general population do not support the accurate prediction of menopause. The ability to predict pregnancy in infertility treatment and natural conception is poor, while a nomogram integrating serum AMH as a stimulation protocol is useful for avoiding poor and/or hyper-responses. On the other hand, improvements in measuring very low concentrations of serum AMH may be capable of distinguishing women with poor ovarian function. Age-independent standardization of AMH values may be helpful for comparing ovarian reserves among women at different ages.


Asunto(s)
Hormona Antimülleriana/sangre , Biomarcadores/sangre , Reserva Ovárica/fisiología , Femenino , Humanos
9.
J Assist Reprod Genet ; 35(2): 339-343, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29080194

RESUMEN

PURPOSE: To visualize and analyze follicle development in ovarian tissue culture using physiological concentrations of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) in order to establish an ovarian tissue culture system that enables efficient in vitro growth of follicles. METHODS: Ovarian tissues from 4-week-old female ICR mice were sliced and cultured. Images of ovarian tissues in culture were obtained at 24-h or 30-min intervals by using a microscope. The area of each follicle observed in the ovarian tissue slices was tracked and analyzed in association with oocyte maturation. RESULTS: We were able to track the development of each follicle using this culture system. Follicle growth was associated with oocyte maturation. Meiotically matured oocytes (MII) were obtained from 33% of all follicles investigated. Approximately, a quarter of follicles (24%) did not grow and resulted in atresia. CONCLUSION: Follicle dynamics were successfully visualized and analyzed in murine ovarian tissue culture. We were able to obtain mature oocytes from the fully grown follicles in vitro. This culture system would be helpful for efficient in vitro culturing of ovarian tissues.


Asunto(s)
Folículo Ovárico/citología , Folículo Ovárico/crecimiento & desarrollo , Ovario/citología , Técnicas de Cultivo de Tejidos/métodos , Animales , Femenino , Células de la Granulosa , Hormona Luteinizante/metabolismo , Meiosis , Ratones Endogámicos ICR , Oocitos/fisiología , Folículo Ovárico/fisiología , Imagen de Lapso de Tiempo
10.
Reprod Biol Endocrinol ; 14(1): 72, 2016 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-27793163

RESUMEN

BACKGROUND: Serum anti-Müllerian hormone (AMH) concentration has been used to assess ovarian reserve in patients with endometriosis, especially when endometrioma surgery is involved. Previously, we reported that decreased serum AMH levels after cystectomy for endometriomas can recover to preoperative levels in some cases. In this present study, we assessed the sequential changes in serum AMH levels before and after cystectomy in terms of the state of the mesosalpinx prior to surgery. METHODS: The retrospective cohort study recruited 53 patients from a series of prospective studies conducted from 2009 to 2015. All patients underwent laparoscopic cystectomy for endometriomas. If either mesosalpinx was involved in the endometrioma or adnexal adhesion before cystectomy, the case was defined as 'involved mesosalpinx' (n = 14). If both mesosalpinx remained anatomically correct, the case was classified as 'intact mesosalpinx' (n = 39). Blood samples were obtained from the patients 2 weeks before surgery, and at 1 month and 1 year after surgery to assess serum AMH levels. RESULTS: The serum AMH levels (the involved group vs. the intact group) were 1.92 vs. 0.98 (P = 0.552) preoperatively, 0.59 vs. 1.99 (P = 0.049) at 1 month postoperatively, and 0.48 vs. 2.37 ng/mL (P = 0.007) at 1 year postoperatively. The involved mesosalpinx group showed a further decrease in serum AMH levels at 1 year postoperatively, while serum AMH levels in the intact mesosalpinx group tended to recover. CONCLUSION: These results suggest that pre-existing mesosalpinx disturbance, in combination with adhesiolysis, may be involved in the medium- and long-term decrease in ovarian reserve after endometrioma surgery. A disturbance in ovarian blood supply via the mesosalpinx may underlie this. TRIAL REGISTRATION: UMIN-CTR UMIN000019369 . Retrospectively registered October 15, 2015.


Asunto(s)
Hormona Antimülleriana/sangre , Ligamento Ancho/cirugía , Endometriosis/cirugía , Quistes Ováricos/cirugía , Enfermedades del Ovario/cirugía , Reserva Ovárica , Adulto , Estudios de Casos y Controles , Estudios de Cohortes , Endometriosis/sangre , Femenino , Procedimientos Quirúrgicos Ginecológicos , Humanos , Laparoscopía , Quistes Ováricos/sangre , Enfermedades del Ovario/sangre , Estudios Retrospectivos , Factores de Riesgo , Adherencias Tisulares/cirugía
11.
J Biol Chem ; 289(4): 2331-43, 2014 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-24311789

RESUMEN

Clostridium difficile infection is a serious and highly prevalent nosocomial disease in which the two large, Rho-glucosylating toxins TcdA and TcdB are the main virulence factors. We report for the first time crystal structures revealing how neutralizing and non-neutralizing single-domain antibodies (sdAbs) recognize the receptor-binding domains (RBDs) of TcdA and TcdB. Surprisingly, the complexes formed by two neutralizing antibodies recognizing TcdA do not show direct interference with the previously identified carbohydrate-binding sites, suggesting that neutralization of toxin activity may be mediated by mechanisms distinct from steric blockage of receptor binding. A camelid sdAb complex also reveals the molecular structure of the TcdB RBD for the first time, facilitating the crystallization of a strongly negatively charged protein fragment that has resisted previous attempts at crystallization and structure determination. Electrospray ionization mass spectrometry measurements confirm the stoichiometries of sdAbs observed in the crystal structures. These studies indicate how key epitopes in the RBDs from TcdA and TcdB are recognized by sdAbs, providing molecular insights into toxin structure and function and providing for the first time a basis for the design of highly specific toxin-specific therapeutic and diagnostic agents.


Asunto(s)
Anticuerpos Antibacterianos/química , Proteínas Bacterianas/química , Toxinas Bacterianas/química , Clostridioides difficile/química , Enterotoxinas/química , Epítopos/química , Anticuerpos de Cadena Única/química , Animales , Anticuerpos Antibacterianos/inmunología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/genética , Toxinas Bacterianas/inmunología , Clostridioides difficile/genética , Clostridioides difficile/inmunología , Cristalografía por Rayos X , Enterotoxinas/antagonistas & inhibidores , Enterotoxinas/genética , Enterotoxinas/inmunología , Epítopos/genética , Epítopos/inmunología , Humanos , Estructura Terciaria de Proteína , Anticuerpos de Cadena Única/inmunología , Relación Estructura-Actividad
12.
Front Microbiol ; 14: 1110541, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36778856

RESUMEN

Crystal structures of camelid heavy-chain antibody variable domains (VHHs) bound to fragments of the combined repetitive oligopeptides domain of Clostridiodes difficile toxin A (TcdA) reveal that the C-terminus of VHH A20 was located 30 Å away from the N-terminus of VHH A26. Based on this observation, we generated a biparatopic fusion protein with A20 at the N-terminus, followed by a (GS)6 linker and A26 at the C-terminus. This A20-A26 fusion protein shows an improvement in binding affinity and a dramatic increase in TcdA neutralization potency (>330-fold [IC 50]; ≥2,700-fold [IC 99]) when compared to the unfused A20 and A26 VHHs. A20-A26 also shows much higher binding affinity and neutralization potency when compared to a series of control antibody constructs that include fusions of two A20 VHHs, fusions of two A26 VHHs, a biparatopic fusion with A26 at the N-terminus and A20 at the C-terminus (A26-A20), and actoxumab. In particular, A20-A26 displays a 310-fold (IC 50) to 29,000-fold (IC 99) higher neutralization potency than A26-A20. Size-exclusion chromatography-multiangle light scattering (SEC-MALS) analyses further reveal that A20-A26 binds to TcdA with 1:1 stoichiometry and simultaneous engagement of both A20 and A26 epitopes as expected based on the biparatopic design inspired by the crystal structures of TcdA bound to A20 and A26. In contrast, the control constructs show varied and heterogeneous binding modes. These results highlight the importance of molecular geometric constraints in generating highly potent antibody-based reagents capable of exploiting the simultaneous binding of more than one paratope to an antigen.

13.
Front Microbiol ; 14: 1167817, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37065148

RESUMEN

[This corrects the article DOI: 10.3389/fmicb.2023.1110541.].

14.
Reprod Sci ; 30(4): 1306-1315, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36194357

RESUMEN

Polycystic ovary syndrome (PCOS), a common endocrine disorder, is associated with impaired oocyte development, leading to infertility. However, the pathogenesis of PCOS has not been completely elucidated. This study aimed to determine the differentially expressed genes (DEGs) and epigenetic changes in the oocytes from a PCOS mouse model to identify the etiological factors. RNA-sequencing analysis revealed that 90 DEGs were upregulated and 27 DEGs were downregulated in mice with PCOS compared with control mice. DNA methylation analysis revealed 30 hypomethylated and 10 hypermethylated regions in the PCOS group. However, the DNA methylation status did not correlate with differential gene expression. The pathway enrichment analysis revealed that five DEGs (Rps21, Rpl36, Rpl36a, Rpl37a, and Rpl22l1) were enriched in ribosome-related pathways in the oocytes of mice with PCOS, and the immunohistochemical analysis revealed significantly upregulated expression levels of Rps21 and Rpl36. These results suggest that differential gene expression in the oocytes of mice in PCOS is related to impaired folliculogenesis. These findings improve our understanding of PCOS pathogenesis.


Asunto(s)
Síndrome del Ovario Poliquístico , Humanos , Femenino , Animales , Ratones , Síndrome del Ovario Poliquístico/metabolismo , Oocitos/metabolismo , Oogénesis/genética , Epigénesis Genética , Perfilación de la Expresión Génica/métodos
15.
Reprod Sci ; 29(12): 3404-3412, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35212933

RESUMEN

Our previous study found that 17ß-estradiol (E2) suppresses primordial follicle activation and growth in cultured mouse ovaries. In this study, we administered tamoxifen, an estrogen receptor antagonist, into the abdominal cavity of mice to clarify the relationship between primordial follicle activation and the physiological concentration of E2 in mouse ovaries. The results showed that tamoxifen promoted primordial follicle activation. Administration of tamoxifen promoted degradation of the extracellular matrix surrounding primordial follicles in the ovaries. Furthermore, tamoxifen decreased the expression of stefin A, an inhibitor of cathepsins that digest some proteins and extracellular matrix, in the ovaries. Mechanical stress produced by the extracellular matrix reportedly suppresses the activation of primordial follicles. The collective results show that tamoxifen can promote primordial follicle activation through the degradation of the extracellular matrix surrounding primordial follicles. Our results indicate that E2 suppresses primordial follicle activation in vivo and that tamoxifen may be useful as a therapeutic agent against infertility.


Asunto(s)
Folículo Ovárico , Tamoxifeno , Femenino , Ratones , Animales , Tamoxifeno/farmacología , Folículo Ovárico/metabolismo , Ovario/metabolismo , Estradiol/farmacología , Estradiol/metabolismo
16.
Cell Death Discov ; 7(1): 186, 2021 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-34285194

RESUMEN

Patients with primary ovarian insufficiency (POI) often have a high prevalence of autoimmune disorders. To identify antigenic molecules associated with ovarian autoimmunity, we performed immunoprecipitation (IP) screening using serum from patients with POI and the established human granulosa cell line (HGrC1). POTE ankyrin domain family member E (POTEE) and POTE ankyrin domain family member F (POTEF), proteins specific to primates, were identified as candidate antigens. Using immunohistochemistry (IHC) with human ovarian tissue, POTEE or POTEF was weakly seen in the granulosa cells (GCs) of primordial follicles and primary follicles, and strongly in large antral follicles and luteal cells. Interestingly, no signals were detected in growing GCs in secondary, preantral, and small antral follicles. Thus, to explore the function of POTEE and POTEF in human folliculogenesis, we established HGrC1 cell lines with drug-inducible expression of POTEF. Expression of POTEF significantly suppressed cell proliferation in HGrC1 cells. Furthermore, chaperonin containing TCP-1 complex (CCT) components, which affect folding proteins required for cell proliferation, was bound to the actin domain of POTEF protein. Although CCT is normally localized only around the Golgi apparatus, TCP-1α, a component of CCT, co-migrated closer to the cell membrane when POTEF expression was induced. These data suggest that the interaction between POTEF and CCT components impairs the usual function of CCT during cell growth. In addition, over-accumulation of POTEF in HGrC1 cells leads to autophagic failure. It was recently reported that knockout of an autophagic gene in mice leads to a phenotype similar to human POI. These results suggested that a proper amount of POTEF is required for the maintenance of GCs in follicle pools, whereas POTEF overaccumulation might be involved in follicle atresia and the development of POI. We also showed the possibility that POTEF could be an antigen involved in ovarian autoimmunity.

17.
Reprod Sci ; 28(1): 31-36, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32737737

RESUMEN

Patients with primary ovarian insufficiency (POI) occasionally present with follicle growth; however, accurately predicting cycles accompanied by follicle growth is challenging. Early-stage follicles produce serum anti-Müllerian hormone (AMH), a useful marker of ovarian reserve. Therefore, serum AMH levels indicate growth of small follicles (which are difficult to detect ultrasonographically) and may predict follicle growth in patients with POI. Using an ultrasensitive enzyme-linked immunosorbent assay (ELISA) kit, we observed very low serum AMH levels in patients with POI. We further evaluated follicle growth in each patient during each cycle to determine the usefulness of measuring serum AMH levels as a predictor of follicle growth in patients with POI who receive hormone replacement therapy (HRT). We investigated 19 patients with POI in whom we analyzed 91 cycles; 14 cycles showed positive and 77 cycles showed negative results on serum AMH testing. The rate of cycles showing follicle growth in AMH-positive cycles was higher than that in AMH-negative cycles (64.3% vs. 6.5%, p = 0.0001). The median serum AMH level (7.7 pg/mL [25th and 75th percentiles 4.6 pg/mL and 22.3 pg/mL, respectively]) in AMH-positive cycles was lower than the lower limit of detection of conventional AMH ELISA kits. The positive predictive value of positive serum AMH levels for follicle growth was higher than that of follicle-stimulating hormone (< 10 mIU/mL). These results indicate that a very low level of serum AMH detected using picoAMH assays is a useful predictor of follicle growth in patients with POI receiving HRT.


Asunto(s)
Hormona Antimülleriana/sangre , Terapia de Reemplazo de Hormonas , Folículo Ovárico/efectos de los fármacos , Insuficiencia Ovárica Primaria/tratamiento farmacológico , Adolescente , Adulto , Biomarcadores/sangre , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Folículo Ovárico/metabolismo , Folículo Ovárico/fisiopatología , Valor Predictivo de las Pruebas , Insuficiencia Ovárica Primaria/sangre , Insuficiencia Ovárica Primaria/diagnóstico , Insuficiencia Ovárica Primaria/fisiopatología , Reproducibilidad de los Resultados , Resultado del Tratamiento , Adulto Joven
18.
F S Sci ; 1(2): 195-205, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35559928

RESUMEN

OBJECTIVE: To establish and characterize cell lines derived from human endometrial epithelial cells (ECs) and mesenchymal cells (MCs) from patients with and without endometriosis. DESIGN: In vitro experimental study. SETTING: University and national cancer center research institute. PATIENT(S): Two women with endometriosis and two women without endometriosis. INTERVENTION(S): Sampling of endometrial ECs and MCs. MAIN OUTCOME MEASURE(S): Establishing immortalized endometrial ECs and MCs with quantitative reverse transcription-polymerase chain reaction (qRT-PCR), immunocytochemical analysis, and RNA sequence profiling performed to characterize the immortalized cells and a cell proliferation assay, three-dimensional culture, and assays for hormone responses performed to characterize the features of ECs. RESULT(S): The qRT-PCR, immunocytochemical analysis, and Western blot analysis revealed that the ECs and MCs maintained their original features. Moreover, the immortalized cells were found to retain responsiveness to sex steroid hormones. The ECs formed a gland-like structure in three-dimensional culture, indicating the maintenance of normal EC phenotypes. The RNA sequence profiling, principal component analysis, and clustering analysis showed that the gene expression patterns of the immortalized cells were different from those of cancer cells. Several signaling pathways that were statistically significantly enriched in ECs and MCs with endometriosis were revealed. CONCLUSION(S): We successfully obtained four paired immortalized endometrial ECs and MCs from patients with and without endometriosis. Using these cells could help identify diagnostic and therapeutic targets for endometriosis. The cell lines established in this study will thus serve as powerful experimental tools in the study of endometriosis.

19.
Reprod Sci ; 27(7): 1400-1410, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32329031

RESUMEN

Endometriosis has several distinguishing features in the ectopic endometrium, including chronic inflammation and fibrosis. According to the retrograde menstruation theory, endometriotic cells are derived from eutopic endometrial cells, and adhesion of endometrial cells to the extracellular matrix can be the initial step in the development of endometriosis. Therefore, we hypothesized that cell adhesion, which mediates a sequence of events in the development of endometriosis triggering inflammatory responses and tissue fibrosis could be a possible therapeutic target for endometriosis. We found co-upregulation of focal adhesion kinase (FAK) and monocyte chemoattractant protein-1 (MCP-1) in the endometriotic tissues compared with that in the normal endometrium. MCP-1 secretion was significantly higher in the endometriotic stromal cells than in the eutopic endometrial stromal cells. Furthermore, co-culture of U937 cells and endometriotic stromal cells upregulated secretion of transforming growth factor-ß1 (TGF-ß1). A FAK inhibitor significantly inhibited the secretion of MCP-1 in the endometriotic stromal cells and TGF-ß1 in the co-culture with macrophages. FAK inhibitor treatment in the murine endometriosis model demonstrated a decrease in the formation of endometriotic lesions as well as the expression of MCP-1 and TGF-ß1. Our results suggest that the FAK-mediated sequential development of endometriosis, including inflammatory response and tissue fibrosis, can be a new therapeutic target in endometriosis.


Asunto(s)
Adhesión Celular/fisiología , Sistemas de Liberación de Medicamentos/métodos , Endometriosis/metabolismo , Endometriosis/patología , Quinasa 1 de Adhesión Focal/metabolismo , Mediadores de Inflamación/metabolismo , Animales , Adhesión Celular/efectos de los fármacos , Quimiocina CCL2/antagonistas & inhibidores , Quimiocina CCL2/metabolismo , Técnicas de Cocultivo , Endometriosis/tratamiento farmacológico , Femenino , Fibrosis , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Humanos , Mediadores de Inflamación/antagonistas & inhibidores , Leiomioma/tratamiento farmacológico , Leiomioma/metabolismo , Leiomioma/patología , Ratones , Ratones Endogámicos BALB C , Quinolonas/administración & dosificación , Sulfonas/administración & dosificación , Células U937
20.
J Vis Exp ; (136)2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29985322

RESUMEN

Mammalian females periodically ovulate an almost constant number of oocytes during each estrus cycle. To sustain such regularity and periodicity, regulation occurs at the hypothalamic-pituitary-gonadal axis level and on developing follicles in the ovary. Despite active studies, follicle development mechanisms are not clear because of the several steps involved from the dormant primordial follicle activation to ovulation, and because of the regulation complexity that differs at each follicular stage. To investigate the mechanisms of follicle development, and the dynamics of follicles throughout the estrus cycle, we developed a mouse ovarian tissue culture model that can be used to observe follicle development using a microscope. Systematic follicle development, periodical ovulation, and follicle atresia can all be reproduced in the cultured ovary model, and the culture conditions can be experimentally modulated. Here, we demonstrate the usefulness of this method in the study of the regulatory mechanisms of follicle development and other ovarian phenomena.


Asunto(s)
Folículo Ovárico/fisiología , Ovario/fisiología , Animales , Femenino , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA