Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 279
Filtrar
1.
BMC Med ; 21(1): 460, 2023 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-37996888

RESUMEN

BACKGROUND: Endometriosis is a common, gynaecological disease characterised by the presence of endometrial-like cells growing outside the uterus. Lesions appear at multiple locations, present with variation in appearance, size and depth of invasion. Despite hormones being the recommended first-line treatment, their efficacy, success and side effects vary widely amongst study populations. Current, hormonal medication for endometriosis is designed to suppress systemic oestrogen. Whether these hormones can influence the lesions themselves is not yet clear. Evidence of hormone receptor expression in endometriotic lesions and their ability to respond is conflicting. A variation in their expression, activation of transcriptional co-regulators and the potential to respond may contribute to their variation in patient outcomes. Identifying patients who would benefit from hormonal treatments remain an important goal in endometriosis research. METHODS: Using gene expression data from endometriosis lesions including endometrioma (OMA, n = 28), superficial peritoneal lesions (SUP, n = 72) and deeply infiltrating lesions (DIE, n = 78), we performed principal component analysis, differential gene expression and gene correlation analyses to assess the impact of menstrual stage, lesion subtype and hormonal treatment on the gene expression. RESULTS: The gene expression profiles did not vary based on menstrual stage, but could distinguish lesion subtypes with OMA significantly differentiating from both SUP and DIE. Additionally, the effect of oestrogen suppression medication altered the gene expression profile in OMA, while such effect was not observed in SUP or DIE. Analysis of the target receptors for hormonal medication indicated ESR2 was differentially expressed in OMA and that genes that correlated with ESR2 varied significantly between medicated and non-medicated OMA samples. CONCLUSIONS: Our results demonstrate of the different lesion types OMA present with strongest response to hormonal treatment directly through ESR2. The data suggests that there may be the potential to target treatment options to individual patients based on pre-surgical diagnoses.


Asunto(s)
Endometriosis , Enfermedades Peritoneales , Femenino , Humanos , Endometriosis/tratamiento farmacológico , Endometriosis/genética , Transcriptoma , Endometrio/metabolismo , Endometrio/patología , Enfermedades Peritoneales/metabolismo , Enfermedades Peritoneales/patología , Estrógenos/metabolismo , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo
2.
Sci Rep ; 12(1): 504, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35017615

RESUMEN

We provide a descriptive characterization of the unfolded protein response (UPR) in skeletal muscle of human patients with peritoneal sepsis and a sepsis model of C57BL/6J mice. Patients undergoing open surgery were included in a cross-sectional study and blood and skeletal muscle samples were taken. Key markers of the UPR and cluster of differentiation 68 (CD68) as surrogate of inflammatory injury were evaluated by real-time PCR and histochemical staining. CD68 mRNA increased with sepsis in skeletal muscle of patients and animals (p < 0.05). Mainly the inositol-requiring enzyme 1α branch of the UPR was upregulated as shown by elevated X-box binding-protein 1 (XBP1u) and its spliced isoform (XBP1s) mRNA (p < 0.05, respectively). Increased expression of Gadd34 indicated activation of PRKR-Like Endoplasmic Reticulum Kinase (PERK) branch of the UPR, and was only observed in mice (p < 0.001) but not human study subjects. Selected cell death signals were upregulated in human and murine muscle, demonstrated by increased bcl-2 associated X protein mRNA and TUNEL staining (p < 0.05). In conclusion we provide a first characterization of the UPR in skeletal muscle in human sepsis.


Asunto(s)
Estrés del Retículo Endoplásmico , Músculo Esquelético/metabolismo , Enfermedades Peritoneales/fisiopatología , Sepsis/fisiopatología , Respuesta de Proteína Desplegada , Anciano , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/genética , Antígenos de Diferenciación Mielomonocítica/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Enfermedades Peritoneales/genética , Enfermedades Peritoneales/metabolismo , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/metabolismo , Sepsis/genética , Sepsis/metabolismo , Proteína 1 de Unión a la X-Box/genética , Proteína 1 de Unión a la X-Box/metabolismo
3.
Int J Surg Pathol ; 30(1): 46-49, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33939556

RESUMEN

Keratin granulomas in the peritoneum are a rare finding with multiple etiologies and can be especially challenging for both the pathologist and the surgeon when these lesions are grossly visible. We report a case of a unique frozen section diagnostic scenario of evaluation of keratin granulomas in the peritoneum of a 47-year-old woman in the setting of multiple potential culprits: endometrial endometrioid adenocarcinoma following fertility sparing treatment, and a concurrent dermoid cyst. We discuss the various etiologies of keratin granulomas in the peritoneum, mechanism of their formation, diagnostic significance, as well as implications of fertility sparing treatments. To the best of our knowledge, this is the only case of keratin granulomas in the peritoneum with multiple distinct potential pathologic culprits as well the only case following fertility sparing treatment.


Asunto(s)
Carcinoma Endometrioide/patología , Quiste Dermoide/patología , Neoplasias Endometriales/patología , Granuloma/patología , Queratinas/metabolismo , Neoplasias Ováricas/patología , Enfermedades Peritoneales/patología , Biomarcadores/metabolismo , Carcinoma Endometrioide/complicaciones , Carcinoma Endometrioide/diagnóstico , Carcinoma Endometrioide/metabolismo , Quiste Dermoide/complicaciones , Quiste Dermoide/diagnóstico , Quiste Dermoide/metabolismo , Diagnóstico Diferencial , Neoplasias Endometriales/complicaciones , Neoplasias Endometriales/diagnóstico , Neoplasias Endometriales/metabolismo , Femenino , Secciones por Congelación , Granuloma/diagnóstico , Granuloma/etiología , Granuloma/metabolismo , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/complicaciones , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/metabolismo , Enfermedades Peritoneales/diagnóstico , Enfermedades Peritoneales/etiología , Enfermedades Peritoneales/metabolismo
4.
J Leukoc Biol ; 111(2): 337-353, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34612523

RESUMEN

The peritoneal cavity, a fluid-containing potential space surrounding the abdominal and pelvic organs, is home to a rich network of immune cells that maintain tissue homeostasis and provide protection against infection. However, under pathological conditions such as peritonitis, endometriosis, and peritoneal carcinomatosis, the peritoneal immune system can become dysregulated, resulting in nonresolving inflammation and disease progression. An enhanced understanding of the factors that regulate peritoneal immune cells under both homeostatic conditions and in disease contexts is therefore required to identify new treatment strategies for these often life-limiting peritoneal pathologies. Type I interferons (T1IFNs) are a family of cytokines with broad immunoregulatory functions, which provide defense against viruses, bacteria, and cancer. There have been numerous reports of immunoregulation by T1IFNs within the peritoneal cavity, which can contribute to both the resolution or propagation of peritoneal disease states, depending on the specifics of the disease setting and local environment. In this review, we provide an overview of the major immune cell populations that reside in the peritoneal cavity (or infiltrate it under inflammatory conditions) and highlight their contribution to the initiation, progression, or resolution of peritoneal diseases. Additionally, we will discuss the role of T1IFNs in the regulation of peritoneal immune cells, and summarize the results of laboratory studies and clinical trials which have investigated T1IFNs in peritonitis/sepsis, endometriosis, and peritoneal carcinomatosis.


Asunto(s)
Inmunidad Celular , Inflamación/inmunología , Interferón Tipo I/farmacología , Cavidad Peritoneal/fisiopatología , Enfermedades Peritoneales/inmunología , Animales , Antivirales/farmacología , Humanos , Inflamación/metabolismo , Inflamación/patología , Inflamación/prevención & control , Enfermedades Peritoneales/metabolismo , Enfermedades Peritoneales/patología , Enfermedades Peritoneales/prevención & control
5.
Front Endocrinol (Lausanne) ; 12: 779183, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867826

RESUMEN

More than 10% of women suffer from endometriosis (EMT) during their reproductive years. EMT can cause pain and infertility and requires further study from multiple perspectives. Previous reports have indicated that an increase inapolipoprotein E (ApoE) may be associated with a lower number of retrieved mature oocytes in older women, and an association between ApoE and spontaneous pregnancy loss may exist in patients with EMT. The purpose of this study was to investigate the existence of an increase in ApoE in follicular fluid (FF) and the possible relationship between ApoE and EMT in Chinese women. In the current study, 217 Chinese women (111 control subjects and 106 EMT patients) were included. The ApoE genotypes were identified by Sanger sequencing. We found that ApoE expression in FF was higher in patients with EMT than in the control group. In addition, a significant difference in ApoE4 carriers (ϵ3/ϵ4, ϵ4/ϵ4) was found between the control subjects and the patients with EMT. Furthermore, a nonparametric test revealed significant differences in the numbers of blastocysts and high-quality blastocysts, but not the hormone levels of FSH, LH, and E2, between the two groups. We also established a multifactor (BMI, high-quality blastocysts, and ϵ4) prediction model with good sensitivity for identifying patients who may suffer from EMT. Our results demonstrate that ApoE expression in FF is increased in EMT, the ApoE-ϵ4 allele is significantly linked to EMT, and a combined analysis of three factors (BMI, high-quality blastocysts, and ϵ4) could be used as a predictor of EMT.


Asunto(s)
Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Endometriosis , Líquido Folicular/metabolismo , Enfermedades Peritoneales , Adulto , Estudios de Casos y Controles , Recuento de Células , China/epidemiología , Endometriosis/epidemiología , Endometriosis/genética , Endometriosis/metabolismo , Femenino , Estudios de Asociación Genética , Genotipo , Humanos , Infertilidad Femenina/diagnóstico , Infertilidad Femenina/genética , Infertilidad Femenina/metabolismo , Infertilidad Femenina/patología , Recuperación del Oocito , Oocitos , Reserva Ovárica/genética , Enfermedades Peritoneales/epidemiología , Enfermedades Peritoneales/genética , Enfermedades Peritoneales/metabolismo , Pronóstico , Regulación hacia Arriba/genética , Adulto Joven
6.
Front Immunol ; 12: 693118, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34489939

RESUMEN

The complement system is a major component of humoral innate immunity, acting as a first line of defense against microbes via opsonization and lysis of pathogens. However, novel roles of the complement system in inflammatory and immunological processes, including in cancer, are emerging. Endometriosis (EM), a benign disease characterized by ectopic endometrial implants, shows certain unique features of cancer, such as the capacity to invade surrounding tissues, and in severe cases, metastatic properties. A defective immune surveillance against autologous tissue deposited in the peritoneal cavity allows immune escape for endometriotic lesions. There is evidence that the glandular epithelial cells found in endometriotic implants produce and secrete the complement component C3. Here, we show, using immunofluorescence and RT-qPCR, the presence of locally synthesized C3 in the ectopic endometriotic tissue, but not in the eutopic tissue. We generated a murine model of EM via injection of minced uterine tissue from a donor mouse into the peritoneum of recipient mice. The wild type mice showed greater amount of cyst formation in the peritoneum compared to C3 knock-out mice. Peritoneal washings from the wild type mice with EM showed more degranulated mast cells compared to C3 knock-out mice, consistent with higher C3a levels in the peritoneal fluid of EM patients. We provide evidence that C3a participates in an auto-amplifying loop leading to mast cell infiltration and activation, which is pathogenic in EM. Thus, C3 can be considered a marker of EM and its local synthesis can promote the engraftment of the endometriotic cysts.


Asunto(s)
Degranulación de la Célula , Complemento C3/metabolismo , Endometriosis/metabolismo , Endometrio/metabolismo , Mediadores de Inflamación/metabolismo , Mastocitos/metabolismo , Enfermedades Peritoneales/metabolismo , Animales , Estudios de Casos y Controles , Técnicas de Cocultivo , Complemento C3/genética , Complemento C3a/metabolismo , Modelos Animales de Enfermedad , Endometriosis/genética , Endometriosis/inmunología , Endometrio/efectos de los fármacos , Endometrio/inmunología , Endometrio/trasplante , Femenino , Células Hep G2 , Humanos , Inmunidad Humoral , Inmunidad Innata , Mastocitos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades Peritoneales/genética , Enfermedades Peritoneales/inmunología , Transducción de Señal , Células THP-1 , Factor de Necrosis Tumoral alfa/farmacología
7.
Reprod Biomed Online ; 43(3): 370-378, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34272164

RESUMEN

Endometriosis is a chronic oestrogen-dependent gynaecological disorder characterized by non-menstrual pelvic pain, infertility and the extrauterine growth of endometrial-like glands and stroma. It has been noted that the eutopic endometrium of women with endometriosis is functionally distinct from that of women without endometriosis. Moreover, ectopic endometrial implants are functionally different from the eutopic endometrium of women with endometriosis. However, the mechanisms directing these differences are ill-defined. It is proposed here that small membrane-bound extracellular vesicles called exosomes are important vehicles in the protection and transport of signalling molecules central to the dysregulation of endometrial function in women with endometriosis. Therefore, a critical review of the literature linking exosomes and their cargo to the pathobiology of endometriosis was conducted. Circulating peritoneal fluid and endometrial cell exosomes contained long non-coding RNA, miRNA and proteins involved in histone modification, angiogenesis and immune modulation that differed significantly in women with endometriosis compared with controls. Moreover, experimental evidence supports a role for exosomes and their cargo in angiogenesis, neurogenesis, immune modulation and endometrial stromal cell invasion. It is therefore suggested that exosomes play an important role in the pathophysiology of endometriosis.


Asunto(s)
Endometriosis/fisiopatología , Endometrio/fisiología , Exosomas/fisiología , Enfermedades Peritoneales/fisiopatología , Endometriosis/genética , Endometriosis/metabolismo , Endometriosis/patología , Endometrio/citología , Células Epiteliales/fisiología , Exosomas/metabolismo , Femenino , Humanos , Sistema Inmunológico/fisiopatología , MicroARNs/metabolismo , MicroARNs/fisiología , Neovascularización Patológica/fisiopatología , Enfermedades Peritoneales/genética , Enfermedades Peritoneales/metabolismo , Enfermedades Peritoneales/patología , Proteínas/metabolismo , Proteínas/fisiología , ARN no Traducido/metabolismo , ARN no Traducido/fisiología , Células del Estroma/fisiología
8.
Reprod Biomed Online ; 43(2): 319-328, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34103260

RESUMEN

RESEARCH QUESTION: What is the potential role of immune cells and their inflammatory cytokines in the pathogenesis, development and establishment of endometriosis? DESIGN: Peritoneal fluid from 59 women (43 with endometriosis and 16 controls) who had undergone laparoscopic surgery was analysed. Changes in the population of innate and adaptive immune cells, cytokines, chemokines and growth factor expression were measured by flow cytometry, Luminex Technology and enzyme-linked immunosorbent assay. RESULTS: No differences were found in the frequencies of the innate and adaptive immune cells between women with and without endometriosis. In the peritoneal fluid of women with endometriosis, IL-1ß, IL-1RN, IL-2, IL-4, IL-8, IL-10, IL-12 (p70), IL-17α, FGF2, G-CSF, MCP-1, MIP-1α and TNF-α were significantly increased compared with controls. A correlation between IL-2, MCP-1, MIP-1α, TNF-α and the severity of endometriosis was observed. The concentration of neopterin, a possible biomarker for this disease, was increased in women with endometriosis compared with controls. CONCLUSIONS: The functional activity of immune cells seemed to be reduced despite their numbers remaining unchanged. The data indicate that a shift of TH cytokine profile occurs, which increases the TH1-TH2 ratio. This is driven by the increased levels of the cytokines (TNF-α and IL-2) in women with severe endometriosis.


Asunto(s)
Endometriosis/inmunología , Tolerancia Inmunológica/fisiología , Enfermedades Peritoneales/inmunología , Adolescente , Adulto , Líquido Ascítico/inmunología , Líquido Ascítico/metabolismo , Líquido Ascítico/patología , Estudios de Casos y Controles , Quimiocinas/metabolismo , Citocinas/metabolismo , Endometriosis/metabolismo , Endometriosis/patología , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Asesinas Naturales/patología , Células Asesinas Naturales/fisiología , Leucocitos/patología , Leucocitos/fisiología , Enfermedades Peritoneales/metabolismo , Enfermedades Peritoneales/patología , Transducción de Señal/inmunología , Adulto Joven
9.
J Clin Endocrinol Metab ; 106(5): 1516-1529, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33507273

RESUMEN

CONTEXT: Small extracellular vesicles (sEVs) have emerged as modulators of the disease microenvironment, thereby supporting disease progression. However, the potential role of EVs and their content to the pathophysiology of endometriosis remain unclear. OBJECTIVE: This work aimed to investigate whether the EVs from eutopic (Eu) and ectopic (Ec) endometrial stromal cells (ESCs) differ with respect to protein composition and role in endometriosis. METHODS: Human Eu and Ec endometrium-derived ESCs were isolated from samples of the same patients (n = 3). sEVs were isolated from ESCs via ultracentrifugation; these sEVs were characterized by Western blotting, transmission electron microscopy, and nanoparticle tracking analysis and analyzed using mass spectrometry. The potential role of EcESCs-derived sEVs (EcESCs-sEVs) in endometriosis was explored by assaying their effects on cell viability/proliferation, migration, and angiogenesis. RESULTS: In total, 105 ESCs-sEV-associated proteins were identified from EcESCs-sEVs and EuESCs-sEVs by mass spectrometry analysis. The protein content differed between EcESCs-sEVs and EuESCs-sEVs, with annexin A2 (ANXA2) being the most prominent difference-present in EcESCs-sEVs but not EuESCs-sEVs. We also found that sEVs-ANXA2 regulates the motility, proliferation, and angiogenesis of ESCs via the extracellularly regulated kinase (ERK)/STAT3 pathway. Notably, treatment of ESCs with sEVs-ANXA2 resulted in increased proliferation and motility, suggesting that sEVs-ANXA2 may be involved in regulating endometriosis. Our data suggest that EcESCs-sEVs-ANXA2 regulates the motility and the angiogenic potential of ESCs, implying a role for sEVs-ANXA2 in the pathogenesis of endometriosis. CONCLUSION: The study of sEVs-ANXA2 from Ec endometriotic cells uncovers a new mechanism of endometriosis progression and will inform the development of novel therapeutic strategies.


Asunto(s)
Endometrio/metabolismo , Vesículas Extracelulares/metabolismo , Células del Estroma/metabolismo , Anexina A2/metabolismo , Anexina A2/fisiología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Micropartículas Derivadas de Células/metabolismo , Células Cultivadas , Endometriosis/metabolismo , Endometriosis/patología , Endometrio/irrigación sanguínea , Endometrio/citología , Vesículas Extracelulares/patología , Femenino , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Enfermedades Peritoneales/metabolismo , Enfermedades Peritoneales/patología , Proteómica , Células del Estroma/citología
10.
J Nucl Med ; 62(1): 69-72, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32444368

RESUMEN

Although radiation-induced mesenteritis or peritonitis can potentially exacerbate the risk of bowel obstruction, there are no data in the literature on the incidence of intestinal obstruction related to peptide receptor radionuclide therapy. Methods: The records of all patients treated with 177Lu-DOTATATE at Moffitt Cancer Center between April 2018 and October 2019 were evaluated. The number of patients who developed bowel obstruction within 3 mo of a 177Lu-DOTATATE treatment was divided by the total number of patients with preexisting peritoneal or mesenteric disease. Management strategies and outcomes were evaluated. Results: Of a total of 159 patients treated, 81 had baseline mesenteric or peritoneal disease, among whom 5 (6%) experienced at least 1 episode of bowel obstruction within 3 mo of treatment. Two of the patients underwent surgical exploration during obstruction describing a "frozen abdomen." All 5 responded at least temporarily to high-dose corticosteroid treatment and regained bowel function, but 2 patients eventually succumbed to progressive peritoneal disease. Conclusion: Peptide receptor radionuclide therapy can lead to bowel obstruction in patients with mesenteric or peritoneal disease, likely by inducing inflammation. Corticosteroids can potentially play a role in treatment and prophylaxis.


Asunto(s)
Obstrucción Intestinal/etiología , Mesenterio/efectos de la radiación , Enfermedades Peritoneales/radioterapia , Receptores de Péptidos/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Octreótido/efectos adversos , Octreótido/análogos & derivados , Octreótido/uso terapéutico , Compuestos Organometálicos/efectos adversos , Compuestos Organometálicos/uso terapéutico , Enfermedades Peritoneales/metabolismo , Riesgo
11.
Int Immunopharmacol ; 90: 107242, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33307514

RESUMEN

Colonic patches, the counterparts of Peyer's patches in the small intestine, are dynamically regulated lymphoid tissues in the colon that have an important role in defensing against microbial infections. Berberine is an isoquinoline alkaloid extracted from medicinal herbs including Rhizoma coptidis and has long been used for the treatment of infectious gastroenteritis, but its impact on the colonic lymphoid tissues (such as colonic patches) is unknown. In this study, we aimed to investigate whether berberine had any influences on the colonic patches in mice with bacterial infection. The results showed that oral berberine administration in bacterial infected mice substantially enhanced the hypertrophy of colonic patches, which usually possessed the features of two large B-cell follicles with a separate T-cell area. Moreover, the colonic patches displayed follicular dendritic cell networks within the B-cell follicles, indicative of mature colonic patches containing germinal centers. Concomitant with enlarged colonic patches, the cultured colon of infected mice treated with berberine secreted significantly higher levels of interleukin-1ß (IL-1ß), IL-6, TNF-α, and CCL-2, while NLRP3 inhibitor MMC950 or knockout of NLRP3 gene abrogated berberine-induced hypertrophy of colonic patches, suggesting the involvement of the NLRP3 signaling pathway in this process. Functionally, oral administration of berberine ameliorated liver inflammation and improved formed feces in the colon. Altogether, these results indicated that berberine was able to augment the hypertrophy of colonic patches in mice with bacterial infection probably through enhancing local inflammatory responses in the colon.


Asunto(s)
Infecciones Bacterianas/patología , Berberina/uso terapéutico , Colon/efectos de los fármacos , Tejido Linfoide/efectos de los fármacos , Enfermedades Peritoneales/patología , Animales , Linfocitos B/efectos de los fármacos , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/metabolismo , Colon/crecimiento & desarrollo , Colon/patología , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Femenino , Gastroenteritis/tratamiento farmacológico , Tejido Linfoide/crecimiento & desarrollo , Tejido Linfoide/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Enfermedades Peritoneales/tratamiento farmacológico , Enfermedades Peritoneales/metabolismo , Linfocitos T/efectos de los fármacos
12.
Gynecol Endocrinol ; 36(sup1): 7-11, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33305662

RESUMEN

OBJECTIVE: The present study was to find a pathogenic evidence for dopamine agonist application in patients with endometriosis associated pain syndrome. PATIENTS AND TECHNIQUE: The study involved 227 patients of reproductive age with histologically confirmed genital endometriosis (GE) of I-III degree according to ASRM classification. The control group included 12 women with no laparoscope detected gynecologic pathology. The levels of prolactin (PRL), peripheral blood (PB), and peritoneal fluid (PF) were evaluated by chemiluminescence immune assay. The pain syndrome was measured by McGill visual analogue scale. Statistica10 program (StatSoft, Inc., Tulsa, OK) was applied for obtained data processing. RESULTS: A correlation was established between GE rate and levels of PRL and PB (Rs = 0.28, p < .05) as well as a correlation of PRL in PB and PF (Rs = 0.29, p < .05). Patients receiving cabergoline combined with hormone therapy standard schemes manifested considerable pain syndrome relief. CONCLUSIONS: PRL involvement in GE pathogenesis and more intense therapeutic impact on pain syndrome in case of combined administration of dopamine and standard hormone therapy prove cabergoline application in clinical practice.


Asunto(s)
Agonistas de Dopamina/uso terapéutico , Endometriosis/tratamiento farmacológico , Enfermedades Peritoneales/tratamiento farmacológico , Adulto , Líquido Ascítico/química , Líquido Ascítico/metabolismo , Cabergolina/uso terapéutico , Quimioterapia Combinada , Endometriosis/complicaciones , Endometriosis/metabolismo , Endometriosis/patología , Femenino , Hormona Liberadora de Gonadotropina/agonistas , Humanos , Terapia Molecular Dirigida/métodos , Dolor Pélvico/tratamiento farmacológico , Dolor Pélvico/etiología , Dolor Pélvico/metabolismo , Enfermedades Peritoneales/complicaciones , Enfermedades Peritoneales/metabolismo , Enfermedades Peritoneales/patología , Prolactina/sangre , Federación de Rusia , Síndrome , Resultado del Tratamiento
13.
Reprod Biomed Online ; 41(6): 1023-1037, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33046374

RESUMEN

RESEARCH QUESTION: Is there a follicular fluid-specific metabolic profile in deep infiltrating endometriosis (DIE) depending on the presence of an associated ovarian endometrioma (OMA) that could lead to the identification of biomarkers for diagnosis and prognosis of the disease? DESIGN: In this prospective cohort study, proton nuclear magnetic resonance (1H-NMR) experiments were carried out on 50 follicular fluid samples from patients presenting with DIE, associated or not associated with an OMA, and 29 follicular fluid samples from patients with infertility caused by a tubal obstruction. RESULTS: Concentrations of glucose, citrate, creatine and amino acids such as tyrosine and alanine were lower in women with DIE than control participants, whereas concentrations of lactate, pyruvate, lipids and ketone bodies were higher. Metabolic analysis revealed enhanced concentrations of glycerol and ketone bodies in patients with OMA, indicative of an activation of lipolysis followed by beta-oxidation. Concentrations of lactate and pyruvate were increased in patients without OMA, whereas the concentration of glucose was decreased, highlighting activation of the anaerobic glycolysis pathway. Differences in concentrations of amino acids such as threonine and glutamine were also statistically relevant in discriminating between the presence or absence of OMA. CONCLUSIONS: Results indicate a mitochondrial dysregulation in endometriosis phenotypes, with a modified balance between anaerobic glycolysis and beta-oxidation in OMA phenotypes that could affect the fertility of women with endometriosis. As the composition of the follicular fluid has been shown to be correlated with oocyte development and outcome of implantation after fertilization, these findings may help explain the high level of infertility in these patients.


Asunto(s)
Endometriosis/metabolismo , Líquido Folicular/metabolismo , Metaboloma , Adulto , Biomarcadores/metabolismo , Estudios de Casos y Controles , Estudios de Cohortes , Endometriosis/clasificación , Endometriosis/patología , Femenino , Líquido Folicular/química , Francia , Humanos , Infertilidad Femenina/etiología , Infertilidad Femenina/metabolismo , Infertilidad Femenina/patología , Metaboloma/fisiología , Persona de Mediana Edad , Enfermedades Peritoneales/clasificación , Enfermedades Peritoneales/metabolismo , Enfermedades Peritoneales/patología , Fenotipo , Estudios Prospectivos
14.
J Clin Endocrinol Metab ; 105(11)2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32813013

RESUMEN

CONTEXT: While fibrosis in endometriosis has recently loomed prominently, the sources of myofibroblasts, the principal effector cell in fibrotic diseases, remain largely obscure. Mesothelial cells (MCs) can be converted into myofibroblasts through mesothelial-mesenchymal transition (MMT) in many fibrotic diseases and adhesion. OBJECTIVE: To evaluate whether MCs contribute to the progression and fibrogenesis in endometriosis through MMT. SETTING, DESIGN, PATIENTS, INTERVENTION, AND MAIN OUTCOME MEASURES: Dual immunofluorescence staining and immunohistochemistry using antibodies against calretinin, Wilms' tumor-1 (WT-1), and α-smooth muscle actin (α-SMA) were performed on lesion samples from 30 patients each with ovarian endometrioma (OE) and deep endometriosis (DE), and 30 normal endometrial (NE) tissue samples. Human pleural and peritoneal MCs were co-cultured with activated platelets or control medium with and without neutralization of transforming growth factor ß1 (TGF-ß1) and/or platelet-derived growth factor receptor (PDGFR) and their morphology, proliferation, and expression levels of genes and proteins known to be involved in MMT were evaluated, along with their migratory and invasive propensity, contractility, and collagen production. RESULTS: The number of calretinin/WT-1 and α-SMA dual-positive fibroblasts in OE/DE lesions was significantly higher than NE samples. The extent of lesional fibrosis correlated positively with the lesional α-SMA staining levels. Human MCs co-cultured with activated platelets acquire a morphology suggestive of MMT, concomitant with increased proliferation, loss of calretinin expression, and marked increase in expression of mesenchymal markers. These changes coincided with functional differentiation as reflected by increased migratory and invasive capacity, contractility, and collagen production. Neutralization of TGF-ß1 and PDGFR signaling abolished platelet-induced MMT in MCs. CONCLUSIONS: MCs contribute to lesional progression and fibrosis through platelet-induced MMT.


Asunto(s)
Plaquetas/metabolismo , Transdiferenciación Celular/fisiología , Endometriosis/patología , Miofibroblastos/patología , Enfermedades del Ovario/patología , Enfermedades Peritoneales/patología , Adulto , Calbindina 2/metabolismo , Línea Celular , Progresión de la Enfermedad , Endometriosis/metabolismo , Femenino , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Persona de Mediana Edad , Miofibroblastos/metabolismo , Enfermedades del Ovario/metabolismo , Enfermedades Peritoneales/metabolismo , Proteínas WT1/metabolismo , Adulto Joven
15.
Int J Mol Sci ; 21(11)2020 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-32532126

RESUMEN

In chronic peritoneal diseases, mesothelial-mesenchymal transition is determined by cues from the extracellular environment rather than just the cellular genome. The transformation of peritoneal mesothelial cells and other host cells into myofibroblasts is mediated by cell membrane receptors, Transforming Growth Factor ß1 (TGF-ß1), Src and Hypoxia-inducible factor (HIF). This article provides a narrative review of the reprogramming of mesothelial mesenchymal transition in chronic peritoneal diseases, drawing on the similarities in pathophysiology between encapsulating peritoneal sclerosis and peritoneal metastasis, with a particular focus on TGF-ß1 signaling and estrogen receptor modulators. Estrogen receptors act at the cell membrane/cytosol as tyrosine kinases that can phosphorylate Src, in a similar way to other receptor tyrosine kinases; or can activate the estrogen response element via nuclear translocation. Tamoxifen can modulate estrogen membrane receptors, and has been shown to be a potent inhibitor of mesothelial-mesenchymal transition (MMT), peritoneal mesothelial cell migration, stromal fibrosis, and neoangiogenesis in the treatment of encapsulating peritoneal sclerosis, with a known side effect and safety profile. The ability of tamoxifen to inhibit the transduction pathways of TGF-ß1 and HIF and achieve a quiescent peritoneal stroma makes it a potential candidate for use in cancer treatments. This is relevant to tumors that spread to the peritoneum, particularly those with mesenchymal phenotypes, such as colorectal CMS4 and MSS/EMT gastric cancers, and pancreatic cancer with its desmoplastic stroma. Morphological changes observed during mesothelial mesenchymal transition can be treated with estrogen receptor modulation and TGF-ß1 inhibition, which may enable the regression of encapsulating peritoneal sclerosis and peritoneal metastasis.


Asunto(s)
Transición Epitelial-Mesenquimal , Moduladores de los Receptores de Estrógeno/farmacología , Enfermedades Peritoneales/tratamiento farmacológico , Enfermedades Peritoneales/patología , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Animales , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Enfermedad Crónica , Células Epiteliales/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Flavonoides/farmacología , Glucólisis/efectos de los fármacos , Glucólisis/fisiología , Humanos , FN-kappa B/metabolismo , Enfermedades Peritoneales/metabolismo , Fibrosis Peritoneal/tratamiento farmacológico , Fibrosis Peritoneal/metabolismo , Fibrosis Peritoneal/patología , Peritoneo/citología , Receptores de Estrógenos/metabolismo , Tamoxifeno/uso terapéutico , Factor de Crecimiento Transformador beta1/metabolismo , Microambiente Tumoral/efectos de los fármacos
16.
Reprod Biol Endocrinol ; 18(1): 63, 2020 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-32532293

RESUMEN

BACKGROUND: A large body of research highlights the importance of early-life environmental impact on the health outcome in adulthood. However, whether early-life adversity (ELA) has any impact on the development of endometriosis is completely unclear. In this study, we tested the hypothesis that ELA, as manifested by neonatal separation, can accelerate the progression of endometriosis in mouse through activation of the adrenergic receptor ß2 (ADRB2) signaling pathway, leading to increased angiogenesis and progression of endometriotic lesions. METHODS: Eight female Balb/C mice, in late pregnancy, were used used for this study, which later gave birth to 22 female newborn pubs. Eleven additional female Balb/C mice were also used as donors of uterine tissues. The 22 newborn pubs were randomly divided into 2 equal-sized groups, maternal separation (MS) and no separation (NS). Pubs in the MS group were separated from their dams for 3 h/day from postnatal day (PND) 1 to 21, while those in the NS control remained in the home cage with their dams. In adulthood (8-week old), 3 mice in each group were randomly selected to undergo a battery of behavior tests. The remaining 8 mice in each group were induced with endometriosis by intraperitoneal injection of uterine fragments from donor mice. Four weeks after the induction, all mice were sacrificed and their endometriotic lesions were excised for quantification and then prepared for immunohistochemistry analysis. RESULTS: We confirmed that MS during infancy resulted in anxiety and depression-like behaviors as previously reported. We also found that in MS mice the lesion weight was increased by over 2 folds and generalized hyperalgesia was also significantly increased as compared with NS mice. Immunostaining analysis demonstrated that MS accelerated the development of endometriosis likely through decreased dopamine receptor D2 (DRD2) expression and activation of the ADRB2/cAMP-response element binding protein (CREB) signaling pathway, leading to increased angiogenesis and progression of endometriotic lesions. CONCLUSIONS: Exposure of female mouse pups to ELA such as MS during their infancy period accelerates the progression of endometriosis, possibly through altered neuronal wiring and hyperactivity of the hypothalamic-pituitary-adrenal axis.


Asunto(s)
Endometriosis , Hiperalgesia , Privación Materna , Enfermedades Peritoneales , Receptores Adrenérgicos beta 2 , Animales , Femenino , Ratones , Animales Recién Nacidos , Ansiedad/psicología , Conducta Animal , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Depresión/psicología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Endometriosis/metabolismo , Endometriosis/patología , Endometriosis/fisiopatología , Endometriosis/psicología , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Hiperalgesia/psicología , Sistema Hipotálamo-Hipofisario/metabolismo , Inyecciones Intraperitoneales , Ratones Endogámicos BALB C , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neovascularización Patológica/fisiopatología , Enfermedades Peritoneales/metabolismo , Enfermedades Peritoneales/patología , Enfermedades Peritoneales/fisiopatología , Enfermedades Peritoneales/psicología , Sistema Hipófiso-Suprarrenal/metabolismo , Distribución Aleatoria , Receptores Adrenérgicos beta 2/metabolismo , Receptores de Dopamina D2/metabolismo , Transducción de Señal , Útero/trasplante , Estrés Psicológico
17.
Hum Reprod Update ; 26(4): 565-585, 2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32412587

RESUMEN

BACKGROUND: Despite intense research, it remains intriguing why hormonal therapies in general and progestins in particular sometimes fail in endometriosis. OBJECTIVE AND RATIONALE: We review here the action mechanisms of progesterone receptor ligands in endometriosis, identify critical differences between the effects of progestins on normal endometrium and endometriosis and envisage pathways to escape drug resistance and improve the therapeutic response of endometriotic lesions to such treatments. SEARCH METHODS: We performed a systematic Pubmed search covering articles published since 1958 about the use of progestins, estro-progestins and selective progesterone receptor modulators, to treat endometriosis and its related symptoms. Two reviewers screened the titles and abstracts to select articles for full-text assessment. OUTCOMES: Progesterone receptor signalling leads to down-regulation of estrogen receptors and restrains local estradiol production through interference with aromatase and 17 beta-hydroxysteroid dehydrogenase type 1. Progestins inhibit cell proliferation, inflammation, neovascularisation and neurogenesis in endometriosis. However, progesterone receptor expression is reduced and disrupted in endometriotic lesions, with predominance of the less active isoform (PRA) over the full-length, active isoform (PRB), due to epigenetic abnormalities affecting the PGR gene transcription. Oxidative stress is another mechanism involved in progesterone resistance in endometriosis. Among the molecular targets of progesterone in the normal endometrium that resist progestin action in endometriotic cells are the nuclear transcription factor FOXO1, matrix metalloproteinases, the transmembrane gap junction protein connexin 43 and paracrine regulators of estradiol metabolism. Compared to other phenotypes, deep endometriosis appears to be more resistant to size regression upon medical treatments. Individual genetic characteristics can affect the bioavailability and pharmacodynamics of hormonal drugs used to treat endometriosis and, hence, explain part of the variability in the therapeutic response. WIDER IMPLICATIONS: Medical treatment of endometriosis needs urgent innovation, which should start by deeper understanding of the disease core features and diverse phenotypes and idiosyncrasies, while moving from pure hormonal treatments to drug combinations or novel molecules capable of restoring the various homeostatic mechanisms disrupted by endometriotic lesions.


Asunto(s)
Endometriosis/tratamiento farmacológico , Ligandos , Enfermedades Peritoneales/tratamiento farmacológico , Receptores de Progesterona/agonistas , Endometriosis/epidemiología , Endometriosis/metabolismo , Endometrio/anomalías , Femenino , Fármacos para la Fertilidad Femenina/uso terapéutico , Humanos , Enfermedades Peritoneales/epidemiología , Enfermedades Peritoneales/metabolismo , Progesterona/uso terapéutico , Progestinas/uso terapéutico , Receptores de Progesterona/metabolismo , Resultado del Tratamiento , Enfermedades Uterinas/tratamiento farmacológico
18.
Reprod Biomed Online ; 41(1): 11-18, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32444257

RESUMEN

RESEARCH QUESTION: What are the protein levels and localization of oestrogen receptors (including ERa, ERb and G protein-coupled oestrogen receptor [GPER]) and hypoxia-inducible factor-1alpha (HIF-1a) in normal control endometrium (COEM) and ectopic endometrium from abdominal wall endometriosis (AWE). DESIGN: AWE (n = 20) were obtained during surgery; COEM (n = 40) were collected by curettage. All tissues were obtained during the proliferative or secretory phase. Formalin-fixed paraffin-embedded tissues were used for immunohistochemical study for oestrogen receptors and HIF-1a proteins. RESULT(S): The expression of oestrogen receptors and HIF-1a in AWE differed from that in the corresponding menstrual cycle phase of COEM. Compared with COEM, ERa and HIF-1a were decreased whereas ERb and GPER were increased in AWE. The greatest difference was in GPER, with increased protein expression in both the cytoplasm and nucleus of endometrial epithelial and stromal cells, as well as a distinct change in localization from cytoplasmic expression to nuclear and cytoplasmic expression, compared with COEM. CONCLUSIONS: Our data suggest that the expression changes of oestrogen receptors and HIF-1a, especially GPER, are associated with AWE, which may provide new clues to understanding the cause of endometriosis.


Asunto(s)
Pared Abdominal , Endometriosis/metabolismo , Endometrio/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Enfermedades Peritoneales/metabolismo , Receptores de Estrógenos/metabolismo , Adulto , Femenino , Humanos , Células del Estroma/metabolismo , Adulto Joven
19.
Mol Cell Endocrinol ; 513: 110870, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32446846

RESUMEN

BACKGROUND: Endometriosis is a benign gynaecological disease with malignant characteristics that severely affects women's quality of life. Long noncoding RNA maternally expressed gene 3 (LncRNA MEG3) is a tumour suppressor that is downregulated in various cancer cells and tissues, and regulates multiple biological processes. Emerging studies have revealed that the interactions between MEG3 and proteins are involved in disease progression. Galectin-1 affects cell motility, signal transduction and vascularization, and is overexpressed in endometriosis. Our study is the first to explore the role of MEG3-210 transcript in endometriosis and to reveal the regulatory mechanism mediated by the interaction between MEG3-210 and Galectin-1. MATERIALS AND METHODS: Endometrial tissues and sera from patients with endometriosis and controls were collected. qRT-PCR was performed to detect the expression of MEG3-210 in the endometrium and endometrial stromal cells (ESCs). The CCK-8 assay, the Transwell assay, flow cytometry and animal models were conducted to evaluate the functions of MEG3-210 in vitro and in vivo. Bioinformatic analysis, Western blot assays, RNA-pull down assays and RNA immunoprecipitation were used to explore the potential mechanism of MEG3-210 in endometriosis. RESULTS: Our results showed that MEG3-210 expression was lower in the eutopic endometrium of women with endometriosis. MEG3-210 downregulation promoted ESCs migration, invasion, anti-apoptosis in vitro and growth of endometriotic lesions in vivo. Furthermore, MEG3-210 downregulation could activate p38 mitogen-activated protein kinase (p38 MAPK) and inhibit cAMP-dependent protein kinase A/sarcoplasmic reticulum Ca2+ ATPase 2 (PKA/SERCA2) signalling, which was mediated by Galectin-1. The protein levels of Galectin-1 in patients with endometriosis were elevated, and Galectin-1 siRNA could reduce the size of lesions. CONCLUSION: MEG3-210 regulates ESCs through p38 MAPK and PKA/SERCA signalling via interaction with Galectin-1. The novel regulatory mechanism may provide new insights into drug therapy and the diagnosis of endometriosis.


Asunto(s)
Endometriosis/genética , Endometrio/fisiología , Galectina 1/metabolismo , Enfermedades Peritoneales/genética , ARN Largo no Codificante/fisiología , Células del Estroma/fisiología , Adulto , Apoptosis/genética , Adhesión Celular/genética , Movimiento Celular/genética , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Endometriosis/metabolismo , Endometriosis/patología , Endometrio/citología , Endometrio/patología , Femenino , Humanos , Enfermedades Peritoneales/metabolismo , Enfermedades Peritoneales/patología , Unión Proteica , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Transducción de Señal/genética , Células del Estroma/patología , Migración Transcelular de la Célula/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
J Clin Endocrinol Metab ; 105(5)2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32119078

RESUMEN

CONTEXT: NOTCH signaling is activated in endometriotic lesions, but the exact mechanisms remains unclear. IL-6, which is increased in the peritoneal fluid of women with endometriosis, induces NOTCH1 through E-proteins including E2A and HEB in cancer. OBJECTIVE: To study the role of E-proteins in inducing NOTCH1 expression under the regulation of IL-6 in endometriosis. SETTING AND DESIGN: The expression of E-proteins and NOTCH1 was first investigated in endometrium of women with endometriosis and the baboon model of endometriosis. Regulation of E-proteins and NOTCH1 expression was examined after IL-6 stimulation and siRNA mediated inhibition of E2A or/and HEB in human endometriotic epithelial cells (12Z) in vitro, and subsequently following IL-6 treatment in the mouse model of endometriosis in vivo. RESULTS: E2A, HEB, and NOTCH1 were significantly upregulated in glandular epithelium (GE) of ectopic endometrium compared to eutopic endometrium in both women and the baboon model. IL-6 treatment upregulated the expression of NOTCH1 together with E2A and HEB in 12Z cells. Small interfering RNA inhibition of E2A and HEB or HEB alone decreased NOTCH1 expression. Binding efficiency of both E2A and HEB was significantly higher at the binding sites on the human NOTCH1 promoter after IL-6 treatment. Finally, IL-6 treatment resulted in a significantly increased number of endometriotic lesions along with increased expression of E2A, HEB, and NOTCH1 in GE of the lesions compared with the vehicle group in an endometriosis mouse model. CONCLUSIONS: IL-6 induced NOTCH1 expression is mediated by E-proteins in the ectopic GE cells, which may promote endometriotic lesion development.


Asunto(s)
Endometriosis/genética , Interleucina-6/farmacología , Enfermedades Peritoneales/genética , Receptor Notch1/genética , Factores de Transcripción/fisiología , Adolescente , Adulto , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Estudios de Casos y Controles , Células Cultivadas , Endometriosis/metabolismo , Endometriosis/patología , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Endometrio/patología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-6/fisiología , Ratones , Persona de Mediana Edad , Papio , Enfermedades Peritoneales/metabolismo , Enfermedades Peritoneales/patología , Receptor Notch1/efectos de los fármacos , Receptor Notch1/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...