Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Reprod Biol Endocrinol ; 22(1): 20, 2024 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-38308254

RESUMEN

BACKGROUND: Decidualization of endometrial cells is the prerequisite for embryo implantation and subsequent placenta formation and is induced by rising progesterone levels following ovulation. One of the hormone receptors contributing to endometrial homeostasis is Progesterone Receptor Membrane Component 1 (PGRMC1), a non-classical membrane-bound progesterone receptor with yet unclear function. In this study, we aimed to investigate how PGRMC1 contributes to human decidualization. METHODS: We first analyzed PGRMC1 expression profile during a regular menstrual cycle in RNA-sequencing datasets. To further explore the function of PGRMC1 in human decidualization, we implemented an inducible decidualization system, which is achieved by culturing two human endometrial stromal cell lines in decidualization-inducing medium containing medroxyprogesterone acetate and 8-Br-cAMP. In our system, we measured PGRMC1 expression during hormone induction as well as decidualization status upon PGRMC1 knockdown at different time points. We further conferred proximity ligation assay to identify PGRMC1 interaction partners. RESULTS: In a regular menstrual cycle, PGRMC1 mRNA expression is gradually decreased from the proliferative phase to the secretory phase. In in vitro experiments, we observed that PGRMC1 expression follows a rise-to-decline pattern, in which its expression level initially increased during the first 6 days after induction (PGRMC1 increasing phase) and decreased in the following days (PGRMC1 decreasing phase). Knockdown of PGRMC1 expression before the induction led to a failed decidualization, while its knockdown after induction did not inhibit decidualization, suggesting that the progestin-induced 'PGRMC1 increasing phase' is essential for normal decidualization. Furthermore, we found that the interactions of prohibitin 1 and prohibitin 2 with PGRMC1 were induced upon progestin treatment. Knocking down each of the prohibitins slowed down the decidualization process compared to the control, suggesting that PGRMC1 cooperates with prohibitins to regulate decidualization. CONCLUSIONS: According to our findings, PGRMC1 expression followed a progestin-induced rise-to-decline expression pattern during human endometrial decidualization process; and the correct execution of this expression program was crucial for successful decidualization. Thereby, the results of our in vitro model explained how PGRMC1 dysregulation during decidualization may present a new perspective on infertility-related diseases.


Asunto(s)
Progesterona , Prohibitinas , Embarazo , Femenino , Humanos , Progesterona/farmacología , Progesterona/metabolismo , Decidua/metabolismo , Receptores de Progesterona/genética , Progestinas/metabolismo , Endometrio/metabolismo , Células del Estroma/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo
2.
J Pharmacol Sci ; 153(4): 188-196, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37973216

RESUMEN

Human endometrial stromal cells (ESCs) undergo differentiation, known as decidualization, and endometrial epithelial cells mature around the embryo implantation stage. In the uterus, cyclooxygenase 2 (COX2), the rate-limiting enzyme that produces prostaglandin E2, is expressed in endometrial stromal and epithelial cells, and promotes decidualization of the former cells. Our recent study demonstrated that progesterone receptor membrane component 1 (PGRMC1) is downregulated during decidualization and may be involved in cellular senescence associated with decidualization via the transcription factor forkhead box protein O1 (FOXO1). Therefore, we investigated the role of PGRMC1 in COX2 expression during differentiation and maturation of endometrial stromal and epithelial cells. Inhibition or knockdown of PGRMC1 significantly enhanced differentiation stimuli-induced COX2 expression in both cell types. However, this COX2 expression was suppressed by FOXO1 knockdown or nuclear factor-kappa B (NF-κB) inhibition. Silencing of COX2 expression inhibited PGRMC1 knockdown-induced expression of decidual markers in ESCs. Thus, PGRMC1 may be linked to FOXO1- and NF-κB-mediated COX2 expression in endometrial cells. Taken together, our data suggest that downregulation of PGRMC1 expression facilitates differentiation of endometrial cells, i.e., decidualization and glandular maturation, via upregulation of COX2 expression.


Asunto(s)
Decidua , FN-kappa B , Femenino , Humanos , AMP Cíclico/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Decidua/metabolismo , Endometrio , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , FN-kappa B/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
3.
J Cell Physiol ; 234(1): 709-720, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-30069867

RESUMEN

Progesterone receptor membrane component 1 (PGRMC1) mediates antimitotic and antiapoptotic actions of progesterone in granulosa cells, which indicates that PGRMC1 may play a key role in maintaining the status of granulosa cells. The current study investigated the effects of progesterone on intracellular signaling involved in differentiation, follicle development, inflammatory responses, and antioxidation, and determined the role of PGRMC1 in these processes. Our results demonstrated that progesterone slowed follicle development and inhibited p-ERK1/2, p-p38, caspase-3, p-NF-κB, and p-IκB-α signals involved in differentiation, steroidogenesis, and inflammatory responses in granulosa cells. Progesterone inhibited the steroidogenic acute regulatory protein and the cholesterol side-chain cleavage enzyme and decreased pregnenolone production. A PGRMC1 inhibitor and a PGRMC1 small interfering RNA ablated these inhibitory effects of progesterone. Interfering with PGRMC1 functions also decreased cellular antioxidative effects induced by an oxidant. These results suggest that PGRMC1 might play a critical role in maintaining the status of granulosa cells and balancing follicle numbers.


Asunto(s)
Células de la Granulosa/citología , Proteínas de la Membrana/genética , Folículo Ovárico/crecimiento & desarrollo , Progesterona/metabolismo , Receptores de Progesterona/genética , Apoptosis/genética , Caspasa 3/genética , Diferenciación Celular/genética , Células Cultivadas , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Células de la Granulosa/metabolismo , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Folículo Ovárico/metabolismo , Receptores de Progesterona/antagonistas & inhibidores , Transducción de Señal/genética
4.
J Reprod Dev ; 63(3): 271-277, 2017 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-28321005

RESUMEN

Follicle growth in the mammalian ovary is coordinately controlled by multiple factors to sustain periodic ovulation. In this study, we investigated the role of progesterone on follicle growth in the mouse ovary. As the concentration of progesterone changes during the estrus cycle, we cultured the sliced mouse ovary in a medium containing 10 ng/ml, 100 ng/ml, and 1 µg/ml progesterone. Progesterone promoted the growth of primordial to primary follicles at 100 ng/ml, while it suppressed the growth of secondary follicles at 1 µg/ml. Follicles at other developmental stages in the cultured ovary were unaffected with different concentrations of progesterone. The number of ovulated oocytes increased in the medium containing 100 ng/ml progesterone but decreased in the presence of 1 µg/ml progesterone. Follicles expressed two types of progesterone receptors, progesterone receptor (PGR) and PGR membrane component 1 (PGRMC1). While PGR shows transient expression on granulosa cells of Graafian follicles, PGRMC1 expresses in granulosa cells of developing follicles. These results suggest that progesterone controls the growth of developing follicles through PGRMC1. Our study shows that the effect of progesterone on ovulation and follicle growth in mouse ovary is dependent on the concentration of progesterone and the follicle stage.


Asunto(s)
Folículo Ovárico/crecimiento & desarrollo , Progesterona/fisiología , Animales , Estradiol/metabolismo , Ciclo Estral/sangre , Femenino , Proteínas de la Membrana/metabolismo , Ratones Endogámicos ICR , Folículo Ovárico/metabolismo , Ovulación , Embarazo , Preñez/sangre , Receptores de Progesterona/metabolismo
5.
Gynecol Endocrinol ; 32(1): 58-60, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26303031

RESUMEN

OBJECTIVE: Our and other studies have pointed on an important role of progesterone receptor membrane component 1 (PGRMC1) in development of breast cancer, especially in hormone therapy. To investigate if PGRMC1 could be used to predict the risk for getting breast cancer, we assessed in tissues of patients with primary invasive breast cancer, if the expression of PGRMC1 may be associated with the expression of estrogen receptor alpha (ERα), progesterone receptor (PR), and ki67. METHODS: Samples from 109 patients with breast cancer between the years 2008 and 2014 were obtained with the patients' consent. Each sample was evaluated for the ERα, PR, Ki67, and PGRMC1 expression by immunohistochemistry using serial sections from the ame paraffin block comparing malignant tissue to benign tissue. RESULTS: Expression of PGRMC1 is increased in tumor area compared with non-cancerous tissue and positively correlates with ERα expression (OR = 1.42 95%CI 1.06-1.91, p = 0.02). No association was obtained between expression of PGRMC1 and PR or Ki67. CONCLUSION: It can be suggested that women with breast epithelium highly expressing PGRMC1 and in interaction with ERα may have an increased risk to develop breast cancer, especially when treated with hormone therapy.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Carcinoma Lobular/metabolismo , Receptor alfa de Estrógeno/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de Progesterona/metabolismo , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , Carcinoma Lobular/patología , Terapia de Reemplazo de Estrógeno , Femenino , Humanos , Inmunohistoquímica , Antígeno Ki-67/metabolismo , Modelos Logísticos , Clasificación del Tumor , Riesgo
6.
Reprod Biomed Online ; 29(5): 640-3, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25246111

RESUMEN

The gene PGRMC1 is highly expressed in the granulose and luteal cells of rodent and primate ovaries. Its role in anti-apoptosis and regulating cell-cycle progression suggests a role in regulating follicle growth. The hypothesis is supported by the study in mice and studies in Sweden. In this study, the coding exons of PGRMC1 were sequenced among 196 Chinese women with premature ovarian failure (POF) and 200 controls, and one novel missense mutation was identified (C.556C>T, p. Pro186Ser) in the POF group and one novel SNP (C.533C>T, p. Trh177Ile) was identified in both groups. The mutation is not considered causative because protein prediction did not indicate a deleterious effect. It is concluded that coding mutations of PGRMC1 do not seem to be a common cause of the disease in Han Chinese women. Future studies in larger cohorts from other ethnic groups are necessary to establish the role of PGRMC1 in POF.


Asunto(s)
Células de la Granulosa/metabolismo , Proteínas de la Membrana/genética , Mutación , Insuficiencia Ovárica Primaria/etnología , Insuficiencia Ovárica Primaria/genética , Receptores de Progesterona/genética , Adulto , Apoptosis , Pueblo Asiatico/genética , Estudios de Casos y Controles , Ciclo Celular , China , Codón , Estudios de Cohortes , Cartilla de ADN , Exones , Femenino , Hormona Folículo Estimulante/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Mutación Missense
7.
J Endocrinol ; 260(2)2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-37965940

RESUMEN

Mononuclear cytotrophoblasts (CTs) differentiate and fuse to form multinuclear syncytiotrophoblasts (STs), which produce human chorionic gonadotropin (hCG) and progesterone to maintain pregnancy. Impaired differentiation and fusion of CTs to form STs are associated with hypertensive disorders of pregnancy and fetal growth restriction. Progesterone receptor membrane component 1 (PGRMC1) is a multifunctional single transmembrane heme-binding protein. We previously demonstrated that downregulation of PGRMC1 promotes endometrial stromal cell differentiation (decidualization). Here, we explored the role of PGRMC1 in trophoblast differentiation and fusion. PGRMC1 expression was lower in STs than in CTs of first-trimester placental tissues. PGRMC1 expression in BeWo cells (a trophoblast-derived choriocarcinoma cell line) decreased upon dibutyryl-cAMP (db-cAMP)-induced differentiation. Both inhibition and knockdown of PGRMC1 stimulated hCG production in the presence of db-cAMP. Furthermore, a quantitative cell fusion assay we developed revealed that inhibition and knockdown of PGRMC1 enhanced db-cAMP-stimulated cell fusion. Peroxisome proliferator-activated receptor γ (PPARγ) agonists decreased PGRMC1 expression and stimulated the cell fusion in BeWo cells. These findings suggest that downregulation of PGRMC1 expression in part through activation of PPARγ during trophoblast differentiation promotes hCG production and cell fusion for formation and maintenance of placental villi during pregnancy.


Asunto(s)
PPAR gamma , Placenta , Humanos , Femenino , Embarazo , Regulación hacia Abajo , PPAR gamma/metabolismo , Placenta/metabolismo , Línea Celular , Gonadotropina Coriónica/farmacología , Trofoblastos/fisiología , Diferenciación Celular/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
8.
J Xenobiot ; 14(2): 575-603, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38804287

RESUMEN

Progesterone receptor membrane component 1 (PGRMC1) is one of few proteins that have been recently described as direct modulators of the activity of human cytochrome P450 enzymes (CYP)s. These enzymes form a superfamily of membrane-bound hemoproteins that metabolize a wide variety of physiological, dietary, environmental, and pharmacological compounds. Modulation of CYP activity impacts the detoxification of xenobiotics as well as endogenous pathways such as steroid and fatty acid metabolism, thus playing a central role in homeostasis. This review is focused on nine main topics that include the most relevant aspects of past and current PGRMC1 research, focusing on its role in CYP-mediated drug metabolism. Firstly, a general overview of the main aspects of xenobiotic metabolism is presented (I), followed by an overview of the role of the CYP enzymatic complex (IIa), a section on human disorders associated with defects in CYP enzyme complex activity (IIb), and a brief account of cytochrome b5 (cyt b5)'s effect on CYP activity (IIc). Subsequently, we present a background overview of the history of the molecular characterization of PGRMC1 (III), regarding its structure, expression, and intracellular location (IIIa), and its heme-binding capability and dimerization (IIIb). The next section reflects the different effects PGRMC1 may have on CYP activity (IV), presenting a description of studies on the direct effects on CYP activity (IVa), and a summary of pathways in which PGRMC1's involvement may indirectly affect CYP activity (IVb). The last section of the review is focused on the current challenges of research on the effect of PGRMC1 on CYP activity (V), presenting some future perspectives of research in the field (VI).

9.
Mol Imaging Biol ; 18(2): 172-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26335282

RESUMEN

PURPOSE: We examined the progesterone receptor membrane component 1 (PGRMC1) protein expression in rat brain cells as the prerequisite step to understand the biology of PGRMC1 in the central nervous system (CNS). We also performed correlation studies between the PGRMC1 protein level and the binding activity of a sigma-2 fluorescent probe, SW120, in order to explore the possibility of using sigma-2 radiotracer of positron emission tomography (PET) to noninvasively image the CNS. PROCEDURES: Embryonic primary neurons, astrocytes, oligodendrocytes, and microglia cells were cultured. Immunocytochemistry, Western blot, and SW120 staining were performed in these cells. RESULTS: The protein expression of PGRMC1 determined by immunocytochemistry and SW120 staining is prominent in neurons and relatively low in astrocytes, oligodendrocytes, and microglia cells. The PGRMC1 expression level correlates with the binding activity of SW120 in rat brain cells. CONCLUSIONS: The sigma-2 receptor PET radiotracer can be potentially used to noninvasively image neuron/synapse densities in the CNS.


Asunto(s)
4-Cloro-7-nitrobenzofurazano/análogos & derivados , Compuestos de Azabiciclo/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Colorantes Fluorescentes/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de Progesterona/metabolismo , Receptores sigma/metabolismo , 4-Cloro-7-nitrobenzofurazano/metabolismo , Animales , Astrocitos/metabolismo , Western Blotting , Células Cultivadas , Hipocampo/metabolismo , Inmunohistoquímica , Microglía/metabolismo , Neuronas/metabolismo , Oligodendroglía/metabolismo , Ratas Sprague-Dawley
10.
Free Radic Biol Med ; 99: 333-344, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27565814

RESUMEN

Heme oxygenase (HO) is a mono-oxygenase utilizing heme and molecular oxygen (O2) as substrates to generate biliverdin-IXα and carbon monoxide (CO). HO-1 is inducible under stress conditions, while HO-2 is constitutive. A balance between heme and CO was shown to regulate cell death and survival in many experimental models. However, direct molecular targets to which CO binds to regulate cellular functions remained to be fully examined. We have revealed novel roles of CO-responsive proteins, cystathionine ß-synthase (CBS) and progesterone receptor membrane component 1 (PGRMC1), in regulating cellular functions. CBS possesses a prosthetic heme that allows CO binding to inhibit the enzyme activity and to regulate H2S generation and/or protein arginine methylation. On the other hand, in response to heme accumulation in cells, PGRMC1 forms a stable dimer through stacking interactions of two protruding heme molecules. Heme-mediated PGRMC1 dimerization is necessary to interact with EGF receptor and cytochromes P450 that determine cell proliferation and xenobiotic metabolism. Furthermore, CO interferes with PGRMC1 dimerization by dissociating the heme stacking, and thus results in modulation of cell responses. This article reviews the intriguing functions of these two proteins in response to inducible and constitutive levels of CO with their pathophysiological implications.


Asunto(s)
Monóxido de Carbono/metabolismo , Cistationina betasintasa/metabolismo , Hemo-Oxigenasa 1/metabolismo , Hemo/metabolismo , Homocistinuria/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de Progesterona/metabolismo , Animales , Monóxido de Carbono/química , Cistationina betasintasa/genética , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulación de la Expresión Génica , Hemo/química , Hemo-Oxigenasa 1/genética , Homocistinuria/genética , Homocistinuria/patología , Humanos , Sulfuro de Hidrógeno/química , Sulfuro de Hidrógeno/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Proteínas de la Membrana/genética , Multimerización de Proteína , Receptores de Progesterona/genética , Transducción de Señal
11.
EBioMedicine ; 2(11): 1806-13, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26870805

RESUMEN

The sigma-2 receptor (S2R) is a potential therapeutic target for cancer and neuronal diseases. However, the identity of the S2R has remained a matter of debate. Historically, the S2R has been defined as (1) a binding site with high affinity to 1,3-di-o-tolylguanidine (DTG) and haloperidol but not to the selective sigma-1 receptor ligand (+)-pentazocine, and (2) a protein of 18-21 kDa, as shown by specific photolabeling with [(3)H]-Azido-DTG and [(125)I]-iodoazido-fenpropimorph ([(125)I]-IAF). Recently, the progesterone receptor membrane component 1 (PGRMC1), a 25 kDa protein, was reported to be the S2R (Nature Communications, 2011, 2:380). To confirm this identification, we created PGRMC1 knockout NSC34 cell lines using the CRISPR/Cas9 technology. We found that in NSC34 cells devoid of or overexpressing PGRMC1, the maximum [(3)H]-DTG binding to the S2R (Bmax) as well as the DTG-protectable [(125)I]-IAF photolabeling of the S2R were similar to those of wild-type control cells. Furthermore, the affinities of DTG and haloperidol for PGRMC1 (KI = 472 µM and 350 µM, respectively), as determined in competition with [(3)H]-progesterone, were more than 3 orders of magnitude lower than those reported for the S2R (20-80 nM). These results clarify that PGRMC1 and the S2R are distinct binding sites expressed by different genes.


Asunto(s)
Sitios de Unión , Proteínas de la Membrana/genética , Receptores de Progesterona/genética , Receptores sigma/genética , Empalme Alternativo , Animales , Secuencia de Bases , Línea Celular , Expresión Génica , Técnicas de Inactivación de Genes , Orden Génico , Vectores Genéticos/genética , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Ratones , Datos de Secuencia Molecular , Progesterona/metabolismo , Unión Proteica , Ratas , Receptores de Progesterona/química , Receptores de Progesterona/metabolismo , Receptores sigma/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda