Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Appl Toxicol ; 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38938153

RESUMEN

As a crucial organ of the female reproductive system, the ovary has both reproductive and endocrine functions. Oxidative stress refers to an increase in intracellular reactive oxygen species (ROS), which play a role in the normal physiological activity of the ovary. However, excessive ROS can cause damage to the ovary. With the advancement of human industrial activities, heavy metal pollution has become increasingly severe. Heavy metals cause oxidative stress through both direct and indirect mechanisms, leading to changes in signal transduction pathways that damage the ovaries. This review aims to outline the adverse effects of oxidative stress on the ovaries triggered by heavy metals such as copper, arsenic, cadmium, mercury, and lead. The detrimental effects of heavy metals on ovaries include follicular atresia and decreased estrogen production in experimental animals, and they also cause premature ovarian insufficiency in women. Additionally, this review discusses the role of antioxidants, provides some treatment methods, summarizes the limitations of current research, and offers perspectives for future research directions.

2.
Acta Biochim Biophys Sin (Shanghai) ; 56(2): 239-254, 2024 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-38243680

RESUMEN

The ovarian surface epithelium (OSE) is a single layer of squamous-to-cuboidal epithelial cells that experience repetitive ovulatory rupture and subsequent repair. However, the characteristics of human immortalized ovarian surface epithelial cells (IOSE80) remain elusive. This study aims to determine whether IOSE80 cells have the characteristics of stem cell proliferation and multilineage differentiation and their application in regenerative medicine. IOSE80 cells are sequenced by high-throughput transcriptome analysis, and 5 sets of public data are used to compare the differences between IOSE80 cells and bone marrow mesenchymal stem cells, pluripotent stem cells, and oocytes in transcriptome profiling. The IOSE80 cells present a cobblestone-like monolayer and express the epithelial cell marker KRT18; the stem cell markers IFITM3, ALDH1A1, and VIM; lowly express stem cell marker LGR5 and germ cell markers DDX4 and DAZL. In addition, the GO terms "regulation of stem cell proliferation", "epithelial cell proliferation", etc., are significantly enriched ( P<0.05). IOSE80 cells have the potential to act as mesenchymal stem cells to differentiate into adipocytes with lipid droplets, osteoblasts, and chondroblasts in vitro. IOSE80 cells express pluripotent stem cell markers, including OCT4, SSEA4, TRA-1-60, and TRA-1-81, and they can be induced into three germ layers in vitro. IOSE80 cells also form oocyte-like cells in vitro and in vivo. In addition, IOSE80 cells exhibit robust proliferation, migration, and ovarian repair functions after in vivo transplantation. This study demonstrates that IOSE80 cells have the characteristics of pluripotent/multipotent stem cells, indicating their important role in tissue engineering and regenerative medicine.


Asunto(s)
Células Madre Mesenquimatosas , Células Madre Pluripotentes , Femenino , Humanos , Ovario , Oocitos , Células Epiteliales , Diferenciación Celular/fisiología , Proteínas de la Membrana , Proteínas de Unión al ARN
3.
Public Health Nurs ; 40(1): 97-104, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36300832

RESUMEN

OBJECTIVE: To explore the clinical effect of the tuberculosis (TB) doctor-nurse integration management model METHODS: This study is a retrospective historical cohort study. The clinical data of 180 patients with TB in our hospital from 2019 to 2020 were analyzed retrospectively. In a control group, 90 cases were treated with the traditional medical care model. An observation group of 90 cases received clinical diagnoses, treatments, and nursing under a doctor-nurse integration management model. Comparative analyses between the two groups were conducted on various aspects, including the awareness level of TB prevention and control, medication compliance and patient satisfaction. Comparisons between the two groups were evaluated using independent-sample t-tests or Chi-squared tests RESULTS: Compared with the control group, the knowledge awareness levels of TB prevention and medication compliance in the observation group were significantly higher (p < .05). The appointment waiting times and hospitalization times in the observation group were significantly lower than in the control group (p < .05). The total average satisfaction score of the patients in the observation group was significantly higher than in the control group (p < .05). Compared with the control group, the patients in the observation group were significantly more satisfied with their nursing methods, operating techniques, psychological techniques, service attitudes, and ward management (p < .05). In addition, in the observation group, medical-nursing relationships and doctor-patient communication were better than in the control group; additionally, the satisfaction of doctors with nursing work was also higher than in the control group, which was a statistically significant difference (p < .05) CONCLUSION: The implementation of an integrated medical-nursing cooperation model for TB will help increase the awareness of health knowledge in patients with TB, improve patient medication compliance and enhance patient satisfaction.


Asunto(s)
Tuberculosis , Humanos , Estudios Retrospectivos , Estudios de Cohortes , Tuberculosis/prevención & control , Cumplimiento de la Medicación , Satisfacción del Paciente
4.
Gynecol Endocrinol ; 38(4): 350-353, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35277108

RESUMEN

Normosmic idiopathic hypogonadotropic hypogonadism (nIHH) is a rare disorder with pubertal delay, normal sense of smell. nIHH with a fibroblast growth factor receptor 1 mutation is much more common in adult males but is rarely reported in females. In addition, the assessment and monitoring of ovarian function in nIHH females has often been ignored. We report a 24-year-old nIHH female with the complaint of primary amenorrhea and delayed secondary sexual traits development. Whole-Exome Sequencing analysis revealed a novel mutation in the third exon of fibroblast growth factor receptor 1 gene (c.289 G > A), which resulted in the replacement of glycine acid with serine. Then the patient was recommended to start with the hormone therapy (HT). After several months of estrogen combined with progesterone replacement, the patient had regular menstruation. The breast development and genital development gradually became Tanner stage 5. Anti-Müllerian hormones (AMHs) were also evaluated and the serum AMH level keeps fluctuating within the normal reference range. We highlight the great variability of fibroblast growth factor receptor 1 mutation phenotypes and the evaluation of ovarian reserve in nIHH, and the hormone replacement therapy is necessary to improve secondary sexual development for patients with nIHH.


Asunto(s)
Hipogonadismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Femenino , Humanos , Hipogonadismo/tratamiento farmacológico , Hipogonadismo/genética , Mutación , Fenotipo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Adulto Joven
5.
BMC Womens Health ; 19(1): 31, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30732650

RESUMEN

BACKGROUND: Post-menopause vaginal haemorrhage is typically related to gynaecological malignancies. Bleeding from vaginal varices rarely occurs, especially in nonpregnant women. Moreover, nonpregnancy-related causes of vaginal varicosities include portal hypertension, especially that caused by liver cirrhosis, pelvic congestion syndrome and Klippel-Trenaunay syndrome or Parkes-Weber syndrome. Here, we report an unusual cause of nonpregnancy-associated vaginal variceal bleeding. CASE PRESENTATION: A 55-year-old postmenopausal woman presented in our outpatient department with complaints of recurrent bloody vaginal discharge. A group of varicose veins and several haemorrhagic spots were found on her vaginal wall during a vaginal speculum examination. Genital cancers were excluded by colposcopy and transvaginal ultrasonography; furthermore, a pelvic arteriovenous fistula was not found on a pelvic computed tomography (CT) scan. However, congenital varicosities and deep arteriovenous shunts were observed in her left leg on arterial angiography. Moreover, vaginal bleeding was improved after resolution of the underlying deep arteriovenous shunts in her left leg. Therefore, congenital arteriovenous shunts and elevated inferior vena cava pressure might be responsible for her recurrent vaginal varicose bleeding. CONCLUSION: Haemorrhage due to vaginal varices is easily detected with a vaginal speculum examination. However, diagnosis and treatment of the original disease are important after bleeding is controlled.


Asunto(s)
Várices Esofágicas y Gástricas/diagnóstico , Várices Esofágicas y Gástricas/tratamiento farmacológico , Terapia de Reemplazo de Hormonas , Síndrome de Klippel-Trenaunay-Weber/diagnóstico , Síndrome de Klippel-Trenaunay-Weber/terapia , Hemorragia Uterina/diagnóstico , Hemorragia Uterina/tratamiento farmacológico , Várices Esofágicas y Gástricas/fisiopatología , Femenino , Humanos , Síndrome de Klippel-Trenaunay-Weber/fisiopatología , Persona de Mediana Edad , Posmenopausia , Tomografía Computarizada por Rayos X , Resultado del Tratamiento , Hemorragia Uterina/fisiopatología
6.
Acta Biochim Biophys Sin (Shanghai) ; 51(8): 845-855, 2019 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-31287492

RESUMEN

Autoimmune ovarian disease (AOD) is considered to be a major cause of premature ovarian failure (POF). The immunomodulatory properties of human amniotic epithelial cells (hAECs) have been studied in many disease models. We previously reported that hAECs restored ovarian function in chemotherapy-induced POF mice, but the immunomodulatory mechanism of hAECs is still unclear. To investigate the effect of hAECs on recipient mice, especially on regulatory Treg cells, hAECs and hAEC-conditioned medium (hAEC-CM) were intravenously injected into AOD mice immunized with zona pellucida protein 3 peptides (pZP3). Ovarian function was evaluated through estrous cycle, hormone secretion, follicle development, and cell apoptosis analysis. Immune cells including CD3, CD4, CD8 and Treg cells in the spleens were tested by flow cytometry. To elucidate the effect of hAEC-CM on macrophage function, inflammation model in vitro was established in RAW264.7 cells induced by lipopolysaccharide (LPS). hAECs and hAEC-CM regulated estrous cycles, promoted follicle development, ameliorated cell apoptosis and fibrosis in ovaries of AOD mice. In addition, hAECs significantly reversed the decrease of pZP3-induced Treg cells in the spleens. In vitro, hAEC-CM significantly inhibited the inflammatory reaction induced by LPS in RAW264.7 cells via up-regulating the expression of M2 macrophage genes. Further study demonstrated that hAEC-secreted transforming growth factor-beta and macrophage inhibitory factor played important roles in the macrophage polarization and migration under inflammatory stimulation. Taken together, hAECs restored ovarian function by up-regulating Treg cells in the spleens and reduced the inflammatory reaction via modulating the activated macrophage function in a paracrine manner in the ovaries of AOD mice.


Asunto(s)
Amnios/citología , Enfermedades Autoinmunes/terapia , Células Epiteliales/citología , Enfermedades del Ovario/terapia , Insuficiencia Ovárica Primaria/terapia , Animales , Apoptosis , Movimiento Celular , Medios de Cultivo Condicionados/química , Modelos Animales de Enfermedad , Femenino , Células de la Granulosa/citología , Humanos , Inmunohistoquímica , Inflamación , Oxidorreductasas Intramoleculares/metabolismo , Lipopolisacáridos/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Macrófagos/metabolismo , Ratones , Células RAW 264.7 , Bazo/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Glicoproteínas de la Zona Pelúcida/metabolismo
7.
Cytotherapy ; 20(10): 1247-1258, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30174233

RESUMEN

BACKGROUND AIMS: The chronic inflammation of autoimmune diseases develops repetitive localized destruction or systemic disorders, represented by Hashimoto's thyroiditis (HT) and Systemic lupus erythematosus (SLE) respectively. Currently, there are no efficient ways to treat these autoimmune diseases. Therefore, it is critically important to explore new therapeutic strategies. The aim of this study was to investigate the therapeutic efficacy of human amniotic epithelial cells (hAECs) in murine models of HT and SLE. METHODS: Experimental autoimmune thyroiditis (EAT) was induced in female CBA/J mice by immunization with porcine thyroglobulin (pTg). hAECs were intravenously administered at different time points during the disease course. MRL-Faslpr mice, a strain with spontaneously occurring SLE, were intravenously administered hAECs when their sera were positive for both anti-nuclear antibodies (ANAs) and anti-double-stranded DNA (anti-dsDNA) antibodies. Two weeks after the last cell transplantation, blood and tissue samples were collected for histological examination and immune system analysis. RESULTS: hAECs prevented lymphocytes infiltration into the thyroid and improved the damage of thyroid follicular in EAT mice. Correspondingly, hAECs administration reduced anti-thyroglobulin antibodies (TGAb), anti-thyroid peroxidase antibodies (TPOAb) and thyroid stimulating hormone (TSH) levels. SLE mice injected with hAECs appeared negative for ANAs and anti-dsDNA antibodies and showed reduced immunoglobulin profiles. Mechanically, hAECs modulated the immune cells balance in EAT and SLE mice, by downregulating the ratios of Th17/Treg cells in both EAT and SLE mice and upregulating the proportion of B10 cells in EAT mice. This was confirmed by in vitro assay, in which hAECs inhibited the activation of EAT mice-derived splenocytes. Moreover, hAECs improved the cytokine environment in both EAT and SLE mice, by suppressing the levels of IL-17A and IFN-γ and enhancing TGF-ß. CONCLUSION: These results demonstrated the immunoregulatory effect of hAECs for inflammation inhibition and injury recovery in HT and SLE murine models. The current study may provide a novel therapeutic strategy for these autoimmune diseases in clinic.


Asunto(s)
Amnios/citología , Células Epiteliales/trasplante , Enfermedad de Hashimoto/terapia , Lupus Eritematoso Sistémico/terapia , Animales , Autoanticuerpos/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Femenino , Enfermedad de Hashimoto/inmunología , Humanos , Lupus Eritematoso Sistémico/inmunología , Ratones Endogámicos CBA , Linfocitos T Reguladores/inmunología , Tiroiditis Autoinmune/etiología , Tiroiditis Autoinmune/terapia , Tirotropina/sangre
8.
Acta Pharmacol Sin ; 39(8): 1305-1316, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29565036

RESUMEN

Human amniotic epithelial cells (hAECs), derived from the innermost layer of the term placenta closest to the fetus, have been shown to be potential seed cells for allogeneic cell therapy. Previous studies have shown a certain therapeutic effect of hAECs. However, no appropriate isolation and culture system for hAECs has been developed for clinical applications. In the present study, we established a serum-free protocol for hAEC isolation and cultivation, in which better cell growth was observed compared with that in a traditional culture system with serum. In addition to specific expression of cell surface markers (CD29, CD166 and CD90), characterization of the biological features of hAECs revealed expression of the pluripotent markers SSEA4, OCT4 and NANOG, which was greater than that in human mesenchymal stem cells, whereas very low levels of HLA-DR and HLA-DQ were detected, suggesting the weak immunogenicity of hAECs. Intriguingly, CD90+ hAECs were identified as a unique population with a powerful immunoregulatory capacity. In a systemic safety evaluation, intravenous administration of hAEC did not result in hemolytic, allergy, toxicity issues or, more importantly, tumorigenicity. Finally, the therapeutic effect of hAECs was demonstrated in mice with radiation-induced damage. The results revealed a novel function of hAECs in systemic injury recovery. Therefore, the current study provides an applicable and safe strategy for hAEC cell therapy administration in the clinical setting.


Asunto(s)
Amnios/citología , Células Epiteliales , Trasplante de Células Madre , Animales , Pruebas de Carcinogenicidad , Células Cultivadas , Medio de Cultivo Libre de Suero , Citocinas/metabolismo , Células Epiteliales/fisiología , Células Epiteliales/trasplante , Femenino , Cobayas , Humanos , Masculino , Ratones Endogámicos ICR , Ratones Endogámicos NOD , Ratones SCID , Embarazo , Cultivo Primario de Células , Traumatismos Experimentales por Radiación/terapia , Ratas Sprague-Dawley , Antígenos Thy-1/metabolismo
9.
Acta Biochim Biophys Sin (Shanghai) ; 50(12): 1227-1235, 2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30395153

RESUMEN

Epithelial ovarian cancer (EOC) is the most fatal gynecologic malignancy due to its late diagnosis and lack of curative therapy. The antimalaria compound artemisinin and its derivatives, such as artesunate (ART) and dihydroartemisinin (DHA), have proven to be potent anticancer drugs and act through various anticancer mechanisms. To identify novel targets of artemisinin derivatives in EOC cells, we investigated the effects of ART and DHA on SKOV3 and primary EOC cell growth via CCK-8 assay. Both ART and DHA inhibited EOC cell growth. A cell cycle distribution analysis showed that ART and DHA caused G2/M cell cycle arrest. Moreover, ART and DHA induced autophagy in EOC cells, whereas autophagy inhibitors reversed the cell growth inhibition and cell cycle arrest induced by ART and DHA. Western blot analysis showed that ART and DHA also suppressed the cell cycle-related NF-κB-signaling pathway in EOC cells. These data suggest that artemisinin derivatives induce autophagy, block the cell cycle, and inhibit cell growth in EOC cells. Our research provides new targets for artemisinin derivatives for EOC treatment.


Asunto(s)
Artemisininas/farmacología , Artesunato/farmacología , Autofagia/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Antimaláricos/farmacología , Autofagia/genética , Proteína 5 Relacionada con la Autofagia/genética , Proteína 5 Relacionada con la Autofagia/metabolismo , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/metabolismo , Carcinoma Epitelial de Ovario/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , FN-kappa B/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Interferencia de ARN , Transducción de Señal/efectos de los fármacos
10.
Reprod Biol Endocrinol ; 15(1): 38, 2017 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-28499456

RESUMEN

Premature ovarian insufficiency (POI) is a typical disorder of amenorrhea lasting for a minimum of 4 months. The typical characteristics comprised of declined estrogen and raised serum concentrations of follicle-stimulating hormone (FSH) in women <40-year-old, primarily originating from iatrogenic factors, karyotypic abnormalities, and genetic factors. However, the etiology of POI remains unknown in approximately 90% of cases. POI could lead to infertility, osteoporosis, cardiovascular disorder, and cognitive dysfunction. MicroRNAs (miRNAs) are a class of endogenous noncoding RNAs (ncRNAs) that can mediate post-translational silencing of the genes involved in the regulation of proliferation, differentiation, apoptosis, development, tumorigenesis, and hematopoiesis. Recently, the regulatory functions of miRNAs in the development of POI have been the topic of intensive research. The present review addresses the association of miRNAs' machinery genes (Dicer, Drosha, and XPO5) with POI and the miRNA expression profiles in the plasma of patients with POI. In addition, several specific miRNAs (miR-23a, miR-27a, miR-22-3p, miR-146a, miR-196a, miR-290-295, miR-423, and miR-608) related to POI are also examined in order to highlight the issues that deserve further investigation. A thorough understanding of the exact regulatory roles of miRNAs is imperative to gain novel insights into the etiology of idiopathic POI and offer new research directions in the field.


Asunto(s)
MicroARNs/fisiología , Insuficiencia Ovárica Primaria/genética , Adulto , Amenorrea/genética , Femenino , Humanos , Persona de Mediana Edad
11.
Acta Biochim Biophys Sin (Shanghai) ; 48(11): 998-1005, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27590065

RESUMEN

Human endometrial mesenchymal stem cells (EnSCs) derived from menstrual blood are a unique stem cell source. Evidence suggests that EnSCs exhibit a multi-lineage potential and have attracted extensive attention in regenerative medicine. However, the potential of EnSCs to differentiate into germline cells in vitro remains unclear. In this study, EnSCs were induced to differentiate into germ cells in a differentiation medium supplemented with 20% human follicular fluid. Our results demonstrated that EnSCs derived from human menstrual blood form oocyte-like cells and express germ cell markers. The induced cell aggregates contained not only oocyte-like structures but also cells expressing follicle stimulating hormone receptor and luteotropic hormone receptor, and produced estrogen and progesterone regulated by gonodatropin, suggesting that granulosa-like and theca-like cells were also induced. We further found that granulosa cells promote the development of oocyte-like cells and activate the induction of blastocyst-like structures derived from EnSCs. In conclusion, EnSCs may potentially represent an in vitro system for the investigation of human folliculogenesis.


Asunto(s)
Células Sanguíneas/citología , Diferenciación Celular , Menstruación , Células Madre Mesenquimatosas/citología , Oocitos/citología , Blastocisto/citología , Femenino , Células Germinativas , Células de la Granulosa/citología , Humanos
12.
Acta Biochim Biophys Sin (Shanghai) ; 48(3): 282-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26819076

RESUMEN

Chemoresistance blocks the efficient treatment of epithelial ovarian cancer, which is the most lethal of all gynecological cancers. Cancer stem cells are believed to be at least partially responsible for the development of chemoresistance. In this study, the effect of cisplatin (CDP) on the enrichment and proliferation of cancer stem-like cells (CSLCs) was investigated, and the underlying mechanisms of action were elucidated. An in vitro anchor-free system was employed to enrich CSLCs from the SKOV3 human epithelial ovarian cancer cell line. Our results showed that treatment with low concentrations of CDP resulted in better-enriched CSLCs, with higher proliferative activities. Low dose of CDP was found to induce the expression of chemokine (C-X-C motif) receptor 4 (CXCR4), which is an important stemness marker in cancer stem cells as well as a promising therapeutic target for ovarian cancer treatment. Results also showed that overexpressed CXCR4 generated chemoresistance. Based on these results, it may be concluded that, at low concentrations, CDP itself contributes to the development of drug resistance. This finding provides novel insight into the mechanisms underlying chemoresistance and has significant therapeutic implications for epithelial ovarian cancer treatment.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Cisplatino/administración & dosificación , Células Madre Neoplásicas/patología , Neoplasias Ováricas/patología , Receptores CXCR4/metabolismo , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Humanos
13.
J Transl Med ; 13: 155, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25964118

RESUMEN

BACKGROUND: Human endometrial mesenchymal stem cells (EnSCs) derived from menstrual blood have mesenchymal stem/stromal cells (MSCs) characteristics and can differentiate into cell types that arise from all three germ layers. We hypothesized that EnSCs may offer promise for restoration of ovarian dysfunction associated with premature ovarian failure/insufficiency (POF/POI). METHODS: Mouse ovaries were injured with busulfan and cyclophosphamide (B/C) to create a damaged ovary mouse model. Transplanted EnSCs were injected into the tail vein of sterilized mice (Chemoablated with EnSCs group; n = 80), or culture medium was injected into the sterilized mice via the tail vein as chemoablated group (n = 80). Non-sterilized mice were untreated controls (n = 80). Overall ovarian function was measured using vaginal smears, live imaging, mating trials and immunohistochemical techniques. RESULTS: EnSCs transplantation increased body weight and improved estrous cyclicity as well as restored fertility in sterilized mice. Migration and localization of GFP-labeled EnSCs as measured by live imaging and immunofluorescent methods indicated that GFP-labeled cells were undetectable 48 h after cell transplantation, but were later detected in and localized to the ovarian stroma. 5'-bromodeoxyuridine (BrdU) and mouse vasa homologue (MVH) protein double-positive cells were immunohistochemically detected in mouse ovaries, and EnSC transplantation reduced depletion of the germline stem cell (GSCs) pool induced by chemotherapy. CONCLUSION: EnSCs derived from menstrual blood, as autologous stem cells, may restore damaged ovarian function and offer a suitable clinical strategy for regenerative medicine.


Asunto(s)
Endometrio/patología , Células Madre Mesenquimatosas/citología , Ovario/fisiología , Insuficiencia Ovárica Primaria/metabolismo , Insuficiencia Ovárica Primaria/terapia , Células Madre/citología , Adulto , Animales , Peso Corporal , Busulfano/química , Diferenciación Celular , Ciclofosfamida/química , Modelos Animales de Enfermedad , Ciclo Estral , Femenino , Citometría de Flujo , Células de la Granulosa/citología , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Trasplante de Células Madre
14.
BMC Cancer ; 15: 956, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26673159

RESUMEN

BACKGROUND: As ovarian cancer stem cells (CSCs) are responsible for tumor initiation, invasion, metastasis, and chemo-resistance, new stratagems that selectively target ovarian CSCs are critically significant. Our previous work have demonstrated that ovarian cancer spheroid cells are tumorigenic and chemo-resistant, and have the properties of ovarian CSCs. Herein, we hypothesized that expressing α-gal epitopes on ovarian spheroid cells may help eliminate CSCs and improve the outcome of therapeutic intervention for ovarian cancer patients. METHODS: Lentivirus-mediated transfer of a pig α(1,3)galactosyltransferase [α1,3GT] enzyme gene into human ovarian cell line SKOV3 cells formed α-gal epitope-expressing cells (SKOV3-gal cells), and then these cells were maintained in a serum-free culture system to form SKOV3-gal spheroid cells. Efficacy of this cell vaccine was demonstrated in α1,3GT knockout mice (α1,3GT KO mice). RESULTS: The antibody titers to α-gal epitopes measured by ELISA were significantly increased in α1,3GT KO mice after immunization with SKOV3-gal spheroid cells. Furthermore, compared with the non-immunized KO mice, the SKOV3 tumors grafted under renal capsules of KO mice immunized with SKOV3-gal spheroid cells grew slower and began to shrink on day 12. Western blot analysis also showed that immunized KO mice can produce effective antibody against certain tumor associated antigens (TAAs) derived from both SKOV3 cells and SKOV3 spheroid cells. The TAAs were further investigated by mass spectrometry and RNA interference (RNAi) technology. The results suggested that antibodies responding to protein c-erbB-2 may be raised in the sera of the mice after immunization with SKOV3-gal spheroid cells. Ultimately, vaccination with SKOV3-gal spheroid cells induced more CD3+CD4+T cells in the spleen of immunized mice than non-immunized KO mice. CONCLUSIONS: The results suggest that vaccination using ovarian cancer stem-like cells engineered to express α-gal epitopes may be a novel strategy for treatment of ovarian cancer.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Galactosiltransferasas/inmunología , Neoplasias Glandulares y Epiteliales/inmunología , Células Madre Neoplásicas/inmunología , Neoplasias Ováricas/inmunología , Animales , Western Blotting , Carcinoma Epitelial de Ovario , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Femenino , Citometría de Flujo , Humanos , Espectrometría de Masas , Ratones , Ratones Noqueados , Ratones Desnudos , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Esferoides Celulares/inmunología , Porcinos , Transfección , Células Tumorales Cultivadas
15.
Tissue Eng Part C Methods ; 30(6): 255-267, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38756098

RESUMEN

Human amniotic epithelial cells (hAECs) are novel and promising therapeutic agents for patients suffering from degenerative diseases. Studies have demonstrated that the therapeutic effects of hAECs mainly depend on their paracrine components. Currently, appropriate pretreatment is a widely confirmed strategy for enhancing the repair potential of stem cells; however, the effect of proinflammatory factor pretreatment on hAECs and their secretome is still unclear. In this study, we used the well-characterized proinflammatory factors tumor necrosis factor alpha (TNF-α) and interferon gamma (IFN-γ) to stimulate hAECs and analyzed the effect of TNF-α and IFN-γ on hAECs, including gene expression profile, paracrine proteins, and microRNAs (miRNAs) in exosomes. Results showed that TNF-α and IFN-γ pretreatment improved the viability of hAECs but inhibited the proliferation of hAECs. TNF-α and IFN-γ pretreatment altered the gene expression profile of hAECs, and upregulated differentially expressed genes were predominantly enriched in biological adhesion, antioxidant activity, and response to IFN-beta. In addition, TNF-α and IFN-γ pretreatment enhanced the paracrine secretion of cytokines by hAECs. The upregulated differentially expressed proteins were mainly enriched in tissue remodeling proteins and cytokine-cytokine receptor. Notably, the expression of miRNAs in exosomes from hAECs was also changed by TNF-α and IFN-γ pretreatment. The target genes of upregulated exosomal miRNAs substantially contributed to the response to stimulus, metabolic pathways, and PI3K-Akt signaling pathway. Our findings improve our understanding of the biological characteristics of hAECs after proinflammatory factor pretreatment and provide novel insights to strengthen and optimize the therapeutic potential of hAECs and their secretome in regenerative medicine.


Asunto(s)
Amnios , Células Epiteliales , Humanos , Amnios/citología , Amnios/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Secretoma , Exosomas/metabolismo , Interferón gamma/farmacología , Interferón gamma/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Proliferación Celular/efectos de los fármacos , MicroARNs/metabolismo , MicroARNs/genética , Supervivencia Celular/efectos de los fármacos , Femenino , Citocinas/metabolismo , Células Cultivadas , Mediadores de Inflamación/metabolismo
16.
Adv Sci (Weinh) ; : e2402393, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39229959

RESUMEN

Sleep deprivation (SD) is observed to adversely affect the reproductive health of women. However, its precise physiological mechanisms remain largely elusive. In this study, using a mouse model of SD, it is demonstrated that SD induces the depletion of ovarian primordial follicles, a phenomenon not attributed to immune-mediated attacks or sympathetic nervous system activation. Rather, the excessive secretion of stress hormones, namely norepinephrine (NE) and epinephrine (E), by overactive adrenal glands, has emerged as a key mediator. The communication pathway mediated by the KIT ligand (KITL)-KIT between granulosa cells and oocytes plays a pivotal role in primordial follicle activation. SD heightened the levels of NE/E that stimulates the activation of the KITL-KIT/PI3K and mTOR signaling cascade in an ß2 adrenergic receptor (ADRB2)-dependent manner, thereby promoting primordial follicle activation and consequent primordial follicle loss in vivo. In vitro experiments further corroborate these observations, revealing that ADRB2 upregulates KITL expression in granulosa cells via the activation of the downstream cAMP/PKA pathway. Together, these results reveal the significant involvement of ADRB2 signaling in the depletion of ovarian primordial follicles under sleep-deprived conditions. Additionally, ADRB2 antagonists are proposed for the treatment or prevention of excessive activation of primordial follicles induced by SD.

17.
EClinicalMedicine ; 74: 102744, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39165278

RESUMEN

Background: Premature ovarian failure (POF) is a prevalent and severe condition that impairs female health but there is currently no effective treatment available to restore ovarian function. Human amniotic epithelial cells (hAECs) exhibit ovarian protection in pre-clinical models. Thus, we conducted a single-arm, phase 1 clinical trial to assess the safety and efficacy of allogenic hAECs in treating POF. Methods: A total of 35 patients received 6 × 107 hAECs via ovarian artery and completed a five-month follow-up from December 30, 2020 to January 31, 2022. The follow-up assessments were conducted at various intervals after hAECs treatment, including one month (Visit-1, V-1), three months (Visit-2, V-2), and five months (Visit-3, V-3) post-treatment. The primary endpoints were incidence of adverse events (AEs), and clinically significant laboratory abnormalities. Secondary endpoints included evaluation of transvaginal ultrasound results, sex hormone levels, Menopausal Quality of Life (MENQOL) questionnaire, as well as reproductive indicators. This trial was registered at www.clinicaltrials.gov as NCT02912104. Findings: No serious AEs were observed throughout the five-month follow-up period. The most common AE was hematoma (7/35, 20.00%), and other AEs include pelvic pain (4/35, 11.43%), fever (2/35, 5.71%), anaphylaxis (2/35, 5.71%), and hepatotoxicity (1/35, 2.86%). After hAECs transplantation (hAECT), significant improvements were observed in the levels of endometrial thickness, left ovarian volume, sex hormones (follicle-stimulating hormone (FSH) and estradiol (E2)), and MENQOL scores in all patients during the five-month follow-up period. Among them, 13 participants (37.14%) experienced spontaneous menstrual bleeding, and 20.00% (7/35) reported more than one regular menstrual bleeding post-hAECT. In this response group, significant improvements were observed in endometrial thickness, left ovarian volume, levels of FSH, E2, anti-Müllerian hormone (AMH), and MENQOL scores one month after hAECT in comparison to pre-hAECT. Interpretation: hAECT via ovarian artery is safe, well-tolerated and temporarily ameliorates endometrial thickness, ovarian size, hormone levels, and menopausal symptoms in POF patients. Further randomized controlled trial of hAECs with longer follow-up period and a larger sample size is warranted. Funding: National Natural Science Foundation of China (No. 82271664), the Interdisciplinary Program of Shanghai Jiao Tong University (YG2022ZD028), the Shanghai Municipal Health Committee (202240345), Shanghai Key Laboratory of Embryo Original Diseases (No. Shelab2022ZD01), Shanghai Municipal Education Commission (No. 20152236), and National Key Research and Development Program of China (No. 2018YFC1004802), Shanghai Clinical Research Center for Cell Therapy, China (No. 23J41900100).

18.
BMC Dev Biol ; 13: 34, 2013 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-24006896

RESUMEN

BACKGROUND: Human amniotic fluid cells (hAFCs) may differentiate into multiple cell lineages and thus have a great potential to become a donor cell source for regenerative medicine. The ability of hAFCs to differentiate into germ cell and oocyte-like cells has been previously documented. Herein we report the potential use of hAFCs to help restore follicles in clinical condition involving premature ovarian failure. RESULTS: Human amniotic fluid was obtained via amniocentesis, yielding a subpopulation of cloned hAFCs that was able to form embryoid bodies (EBs) and differentiate into three embryonic germ layers. Moreover, culture of EBs in medium containing human follicular fluid (HFF) or a germ cell maturation factor cocktail (FAC), expressed germ cells markers such as BLIMP1, STELLA, DAZL, VASA, STRA8, SCP3, SCP1, and GDF9. Furthermore, one cell line was grown from clone cells transfected with lentivirus-GFP and displaying morphological characteristics of mesenchymal cells, had the ability to restore ovarian morphology following cell injection into the ovaries of mice sterilized by intraperitoneal injection of cyclophosphamide and busulphan. Restored ovaries displayed many follicle-enclosed oocytes at all stages of development, but no oocytes or follicles were observed in sterilized mice whose ovaries had been injected with medium only (control). Notably, identification of GFP-labeled cells and immunostaining with anti-human antigen-specific antibodies demonstrated that grafted hAFCs survived and differentiated into granulosa cells which directed oocyte maturation. Furthermore, labeling of ovarian tissue for anti-Müllerian hormone expression, a functional marker of folliculogenesis, was strong in hAFCs-transplanted ovaries but inexistent in negative controls. CONCLUSION: These findings highlight the possibility of using human amniotic fluid-derived stem cells in regenerative medicine, in particular in the area of reproductive health.


Asunto(s)
Líquido Amniótico/citología , Líquido Amniótico/metabolismo , Antineoplásicos/efectos adversos , Ciclofosfamida/efectos adversos , Infertilidad Femenina/terapia , Ovario/fisiología , Células Madre Pluripotentes/fisiología , Trasplante de Células Madre , Animales , Antineoplásicos/administración & dosificación , Diferenciación Celular , Células Cultivadas , Ciclofosfamida/administración & dosificación , Modelos Animales de Enfermedad , Cuerpos Embrioides/citología , Femenino , Humanos , Infertilidad Femenina/inducido químicamente , Cariotipo , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos C57BL , Oocitos/citología , Oocitos/metabolismo , Pruebas de Función Ovárica , Células Madre Pluripotentes/trasplante , Embarazo
19.
Cancer Sci ; 104(12): 1683-9, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24033540

RESUMEN

The important functions of heat shock factor 1 (HSF1) in certain malignant cancers have granted it to be an appealing target for developing novel strategy for cancer therapy. Here, we report that higher HSF1 expression is associated with more aggressive malignization in epithelial ovarian tumors, indicating that targeting HSF1 is also a promising strategy against ovarian cancer. We found that a nucleoside analog (Ly101-4B) elicits efficient inhibition on HSF1 expression and potent anticancer activity on epithelial ovarian cancer both in vitro and in vivo. Moreover, by targeting HSF1, Ly101-4B inhibits the biogenesis of microRNA-214, which has been revealed to be overexpressed and to promote cell survival in human ovarian epithelial tumors. These findings demonstrate that Ly101-4B is a promising candidate for ovarian cancer therapy, and expand our understanding of HSF1, by revealing that it can regulate microRNA biogenesis in addition to its canonical function of regulating protein-coding RNAs.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , MicroARNs/antagonistas & inhibidores , Neoplasias Glandulares y Epiteliales/metabolismo , Nucleósidos/farmacología , Neoplasias Ováricas/metabolismo , Factores de Transcripción/metabolismo , Triazoles/farmacología , Animales , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Femenino , Proteínas de Choque Térmico HSP27/genética , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/genética , Factores de Transcripción del Choque Térmico , Proteínas de Choque Térmico , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Chaperonas Moleculares , Trasplante de Neoplasias , Interferencia de ARN , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
20.
Acta Biochim Biophys Sin (Shanghai) ; 45(11): 928-37, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24028972

RESUMEN

Fibroblast activation protein alpha (FAPα) is a 95-kDa serine protease of post-prolyl peptidase family on cell surface. FAPα is widely expressed in tumor microenvironment. The wide spread association of FAPα expression with cancer suggests that it has important functions in the disease. However, the nature of FAPα's roles in cancer cell activity is not well-determined. It has been showed that FAPα silencing in SKOV3 cells induces ovarian tumors but significantly reduces tumor growth in a xenograft mouse model. To further determine the role of FAPα in epithelial ovarian cancer cells, SKOV3-FAPα and HO8910-FAPα cell lines, which over-expressed FAPα stably, were constructed and then their biological behaviors were investigated. It was found that FAPα promoted ovarian cancer cell proliferation, drug resistance, invasiveness, and migration in vitro. Immunochemistry assay showed that FAPα significantly facilitated tumor growth in xenograft tumor tissues. These results suggested that FAPα might directly promote tumor growth and invasiveness in ovarian cancer cells.


Asunto(s)
Proliferación Celular , Gelatinasas/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias Ováricas/patología , Serina Endopeptidasas/metabolismo , Animales , Secuencia de Bases , Línea Celular Tumoral , Cartilla de ADN , Endopeptidasas , Femenino , Gelatinasas/genética , Silenciador del Gen , Xenoinjertos , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , Neoplasias Ováricas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina Endopeptidasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA