Expression and role of resistin on steroid secretion in the porcine corpus luteum

Reproduction ◽  
2021 ◽  
Author(s):  
Patrycja Kurowska ◽  
Monika Sroka ◽  
Monika Dawid ◽  
Ewa Mlyczyńska ◽  
Natalia Respekta ◽  
...  

Resistin plays an important role in adipogenesis, obesity, insulin resistance and reproduction. Previous studies showed resistin action on ovarian follicular cells; however, whether resistin regulates steroid secretion in luteal cells is still unknown. Our aim was first to determine the expression of resistin and its potential receptors (tyrosine kinase-like orphan receptor 1 [ROR1] and Toll-like receptor 4 [TLR4]) in the porcine corpus luteum (CL), regulation of its expression, effect on kinases phosphorylation and luteal steroidogenesis. Our results showed that the expression of resistin and its receptors was dependent on the luteal phase and this was at the mRNA level higher in the late compared with the early and middle luteal phase. At the opposite, resistin protein expression was higher in the middle and late compared with the early luteal phase, while ROR1 and TLR4 expression was highest in the early luteal phase. Additionally, we observed cytoplasmic localisation of resistin, ROR1 and TLR4 in small and large luteal cells. We found that luteinising hormone, progesterone (P4), insulin and insulin-like growth factor 1 regulated the protein level of resistin, ROR1 and TLR4. Resistin decreased P4 and increased oestradiol (E2) secretion via changing in steroidogenic enzymes expression and via the activation of protein kinase A (PKA) and mitogen-activated protein kinase (MAP3/1), increased the expression of receptors LHCGR and ESR2 and decreased the expression of PGR. Moreover, resistin decreased PKA phosphorylation and enhanced MAP3/1 phosphorylation. Taken together, resistin could act directly on steroid synthesis and serve as an important factor in in vivo luteal cell function.

2019 ◽  
Vol 31 (9) ◽  
pp. 1444
Author(s):  
Di Zhang ◽  
Ying Liu ◽  
Yan Cui ◽  
Sheng Cui

Progesterone (P4) synthesized by the corpus luteum (CL) plays a key role in the establishment and maintenance of pregnancy. The LH signal is important for luteinisation and P4 synthesis in pigs. In a previous study, we demonstrated that mitogen-activated protein kinase kinase kinase 8 (MAP3K8) regulates P4 synthesis in mouse CL, but whether the function and mechanism of MAP3K8 in the pig is similar to that in the mouse is not known. Thus, in the present study we investigated the effects of MAP3K8 on porcine CL. Abundant expression of MAP3K8 was detected in porcine CL, and, in pigs, MAP3K8 expression was higher in mature CLs (or those of the mid-luteal phase) than in regressing CLs (late luteal phase). Further functional studies in cultured porcine luteal cells showed that P4 synthesis and the expression of genes encoding the key enzymes in P4 synthesis are significantly reduced when MAP3K8 is inhibited with the MAP3K8 inhibitor Tpl2 kinase inhibitor (MAP3K8i, 10μM). After 12–24h treatment of luteal cells with 100ngmL−1 LH, MAP3K8 expression and P4 secretion were significantly upregulated. In addition, the 10μM MAP3K8 inhibitor blocked the stimulatory effect of LH on P4 synthesis and extracellular signal-regulated kinase (ERK) 1/2 phosphorylation in porcine luteal cells. The LH-induced increases in MAP3K8 phosphorylation and expression, ERK1/2 phosphorylation and P4 synthesis were all blocked when protein kinase A was inhibited by its inhibitor H89 (20 μM) in porcine luteal cells. In conclusion, MAP3K8 mediates the LH-induced stimulation of P4 synthesis through the PKA/mitogen-activated protein kinase signalling pathway in porcine CL.


1987 ◽  
Vol 40 (3) ◽  
pp. 331 ◽  
Author(s):  
William Hansel ◽  
Hector W Alila ◽  
Joseph P Dowd ◽  
Xiangzhong Yang

Evidence was cited to show that: (1) prostacyclin (PGI2) plays a luteotrophic role in the bovine corpus luteum and that products of the lipoxygenase pathway of arachidonic acid metabolism, especially 5-hydroxyeicosatetraenoic acid play luteolytic roles; (2) oxytocin of luteal cell origin plays a role in development, and possibly in regression, of the bovine corpus luteum; and (3) luteal cells arise from two sources; the characteristic small luteal cells at all stages of the o~strous cycle and pregnancy are of theca cell origin; the large cells are of granulosa cell origin early in the cycle, but a population of theca-derived large cells appears later in the cycle. Results of in vitro studies with total dispersed cells and essentially pure preparations of large and small luteal cells indicate that : (1) the recently described Ca2+ -polyphosphoinositol-protein kinase C second messenger system is involved in progesterone synthesis in the bovine corpus luteum; (2) activation of protein kinase C is stimulatory to progesterone synthesis in the small luteal cells; (3) activation of protein kinase C has no effect on progesterone synthesis in the large luteal cells; and (4) protein kinase C exerts its luteotrophic effect in total cell preparations, in part at least, by stimulating the production of prostacyclin. The protein kinase C system may cause down regulation of LH receptors in the large cells.


Reproduction ◽  
2013 ◽  
Vol 146 (6) ◽  
pp. 527-537 ◽  
Author(s):  
Daniel H Poole ◽  
Kalidou Ndiaye ◽  
Joy L Pate

Secreted phosphoprotein 1 (SPP1) in the bovine corpus luteum (CL) regulates cell function during the transitional periods of luteinization and luteal regression. The objectives were to i) characterize SPP1 expression in the CL throughout the estrous cycle, ii) determine factors that regulate SPP1 expression in luteal cells, and iii) examine the role of SPP1 in lymphocyte chemotaxis, proliferation, and function.SPP1mRNA was greater in fully functional (d10) CL and late cycle (d18) CL compared with developing (d4) CL. Additionally,SPP1mRNA increased within 1 h and remained elevated 4 and 8 h following induction of luteolysis with prostaglandin (PG)F2α. Expression of the SPP1 receptor, β3integrin, was not different throughout the estrous cycle but decreased following induction of luteolysis. Expression ofCD44increased during the estrous cycle but did not change during luteal regression. In cultured luteal cells,SPP1mRNA was upregulated by PGF2αand/or tumor necrosis factor α. Western blots revealed the presence of both full-length SPP1 and multiple cleavage products in cultured luteal cells and luteal tissue. Depletion of endogenous SPP1 did not hinder luteal cell-induced lymphocyte proliferation or lymphocyte phenotype but did inhibit lymphocyte migration toward luteal cells. Based on these data, it is concluded that SPP1 is initially activated to establish and maintain cellular interactions between steroidogenic and nonsteroidogenic cells during the development of the CL. Upon induction of luteolysis, SPP1 serves as a signaling molecule to recruit or activate immune cells to facilitate luteal regression and tissue degradation.


2005 ◽  
Vol 17 (6) ◽  
pp. 659 ◽  
Author(s):  
R. P. Roberto da Costa ◽  
V. Branco ◽  
P. Pessa ◽  
J. Robalo Silva ◽  
G. Ferreira-Dias

Steroid hormones act via specific receptors, and these play an important physiological role in the ovary. The objective of this study was to evaluate the cellular distribution of progesterone receptors and their staining intensity in different equine luteal structures during the breeding season, as well as their relationship to luteal cell composition, cell proliferation pattern and plasma progesterone (P4) concentration. There was an increase in proliferating cell nuclear antigen (PCNA) expression in large luteal cells from the corpus hemorrhagicum (CH) to mid-luteal phase, followed by a decrease toward the late luteal stage. In the CH, the number of large luteal cells was lower than in other structures. Only large luteal cells showed positive staining for P4 receptors. An increase in staining intensity for P4 receptors was observed between CH and mid-phase corpus luteum, and CH and late-phase corpus luteum. Synthesis of P4 started at a very early stage of the luteal structure and was accompanied by an increase in P4 receptors and PCNA expression, and proliferation of large luteal cells, until mid-luteal phase. These data suggest that large luteal cells might play an important role in the regulation or synthesis of P4 in equine luteal structures.


PeerJ ◽  
2019 ◽  
Vol 7 ◽  
pp. e6344
Author(s):  
Sang Hwan Kim ◽  
Ji Hye Lee ◽  
Jong Taek Yoon

Here we investigated the expressions of apoptosis-associated genes known to induce programed cell death through mRNA expressions of two matrix metalloproteinases (MMPs) that are involved in the degradation of collagen and basal membrane in luteal cells cultured in the treatment media. Our results show that the activity of MMP-2 gelatinase was higher in the CL2 and CL1 of luteal phase, was gradually decreased in the CH2 and CH3 of luteal phase. In particular, the expressions of P4-r and survival-associated genes (IGFr, PI3K, AKT, and mTOR) were strongly induced during CL3 stage, whereas the levels of these genes in corpus luteum (CL) were lower during CL2 and CL1 stages. In the cultured lutein cells analyzed, we found that as MMPs increase, genes related to apoptosis (20α-hydroxy steroid dehydrogenase and caspase-3) also increase. In other words, the results for P4-r and survival-related gene expression patterns in the luteal cells were contrary to the MMPs activation results. These results indicate that active MMPs are differentially expressed to induce the expression of genes associated with programed cell death from the degrading luteal cells. Therefore, our results suggest that the MMPs activation may lead to luteal cell development or death.


Author(s):  
Emilia Przygrodzka ◽  
Corrine F. Monaco ◽  
Michele R. Plewes ◽  
Guojuan Li ◽  
Jennifer R. Wood ◽  
...  

In the absence of pregnancy the ovarian corpus luteum undergoes regression, a process characterized by decreased production of progesterone and structural luteolysis involving apoptosis. Autophagy has been observed in the corpus luteum during luteal regression. Autophagy is a self-degradative process important for balancing sources of cellular energy at critical times in development and in response to nutrient stress, but it can also lead to apoptosis. Mechanistic target of rapamycin (MTOR) and 5′ AMP-activated protein kinase (AMPK), key players in autophagy, are known to inhibit or activate autophagy, respectively. Here, we analyzed the signaling pathways regulating the initiation of autophagy in bovine luteal cells. In vivo studies showed increased activating phosphorylation of AMPKα (Thr172) and elevated content of LC3B, a known marker of autophagy, in luteal tissue during PGF2α-induced luteolysis. In vitro, AMPK activators 1) stimulated phosphorylation of regulatory associated protein of MTOR (RPTOR) leading to decreased activity of MTOR, 2) increased phosphorylation of Unc-51-Like Kinase 1 (ULK1) and Beclin 1 (BECN1), at sites specific for AMPK and required for autophagy initiation, 3) increased levels of LC3B, and 4) enhanced colocalization of autophagosomes with lysosomes indicating elevated autophagy. In contrast, LH/PKA signaling in luteal cells 1) reduced activation of AMPKα and phosphorylation of RPTOR, 2) elevated MTOR activity, 3) stimulated phosphorylation of ULK1 at site required for ULK1 inactivation, and 4) inhibited autophagosome formation as reflected by reduced content of LC3B-II. Pretreatment with AICAR, a pharmacological activator of AMPK, inhibited LH-mediated effects on RPTOR, ULK1 and BECN1. Our results indicate that luteotrophic signaling via LH/PKA/MTOR inhibits, while luteolytic signaling via PGF2α/Ca2+/AMPK activates key signaling pathways involved in luteal cell autophagy.


1968 ◽  
Vol 59 (2_Suppl) ◽  
pp. S35-S51 ◽  
Author(s):  
B. L. Lobel ◽  
E. Levy

ABSTRACT Activities of various hydrolases and dehydrogenases were studied during the formation, development and involution of cyclic corpora lutea and in the corpora lutea of early pregnancy. At 24 hours postovulation the luteal cells, whether of granulosal or thecal origin, contained demonstrable levels of Δ5-3β-hydroxysteroid dehydrogenase and the NADP and NADPH2 diaphorases. During the period of proliferation and cellular growth, enzymic activities in the luteal cells were moderate at first, and then increased. In the mature corpus luteum, activities of the dehydrogenases occurred in all luteal cells but were most intense in the large polymorphic luteal cells. Activities of hydrolytic enzymes, low in the immediate postovulatory period, increased with the development of the vascular system. Enzymic characteristics of corpora lutea of gestation were similar to those of cyclic corpora, except for phosphorylase activity which was observed in luteal cells in gestational corpora, but confined to the vascular walls in cyclic corpora. No increase in activities of 17β- and 20β-hydroxysteroid dehydrogenases (above those seen in pre-ovulatory follicles) were observed after incubation of sections of either mature cyclic or gestational corpora. Involution of cyclic corpora lutea began with degenerative changes in the blood vessels: pyknosis of the endothelial cell nuclei and a sudden decline in activities of hydrolytic enzymes in the vascular walls. Subsequently, the luteal cells showed a sharp decrease in activities of the dehydrogenases as well as other signs of regressive change. The cytochemical findings are discussed in relation to biochemical observations on steroid synthesis by the bovine corpus luteum.


2021 ◽  
Vol 2021 ◽  
pp. 1-11
Author(s):  
Zonghao Tang ◽  
Jiajie Chen ◽  
Zhenghong Zhang ◽  
Jingjing Bi ◽  
Renfeng Xu ◽  
...  

The increase of oxidative stress is one of the important characteristics of mammalian luteal regression. Previous investigations have revealed the essential role of reactive oxygen species (ROS) in luteal cell death during luteolysis, while it is unknown how ROS is regulated in this process. Considering the decrease of blood flow and increase of PGF2α during luteolysis, we hypothesized that the HIF-1α pathway may be involved in the regulation of ROS in the luteal cell of the late corpus luteum (CL). Here, by using a pseudopregnant rat model, we showed that the level of both HIF-1α and its downstream BNIP3 was increased during luteal regression. Consistently, we observed the increase of autophagy level during luteolysis, which is regulated in a Beclin1-independent manner. Comparing with early (Day 7 of pseudopregnancy) and middle CL (Day 14), the level of ROS was significantly increased in late CL, indicating the contribution of oxidative stress in luteolysis. Inhibition of HIF-1α by echinomycin (Ech), a potent HIF-1α inhibitor, ameliorated the upregulation of BNIP3 and NIX, as well as the induction of autophagy and the accumulation of ROS in luteal cells on Day 21 of pseudopregnancy. Morphologically, Ech treatment delayed the atrophy of the luteal structure at the late-luteal stage. An in vitro study indicated that inhibition of HIF-1α can also attenuate PGF2α-induced ROS and luteal cell apoptosis. Furthermore, the decrease of cell apoptosis can also be observed by ROS inhibition under PGF2α treatment. Taken together, our results indicated that HIF-1α signaling is involved in the regression of CL by modulating ROS production via orchestrating autophagy. Inhibition of HIF-1α could obviously hamper the apoptosis of luteal cells and the process of luteal regression.


1997 ◽  
Vol 45 (1) ◽  
pp. 71-77 ◽  
Author(s):  
Firyal S. Khan-Dawood ◽  
Jun Yang ◽  
M. Yusoff Dawood

We have recently shown the presence of E-cadherin and of α- and γ-catenins in human and baboon corpora lutea. These are components of adherens junctions between cells. The cytoplasmic catenins link the cell membrane-associated cadherins to the actin-based cytoskeleton. This interaction is necessary for the functional activity of the E-cad-herins. Our aim therefore was to determine the presence of α-actin in the baboon corpus luteum, to further establish whether the necessary components for E-cadherin activity are present in this tissue. An antibody specific for the smooth muscle isoform of actin, α-actin, was used for these studies. The results using immunohistochemistry show that (a) α-actin is present in steroidogenic cells of the active corpus luteum, theca externa of the corpus luteum, cells of the vasculature, and the tunica albuginea surrounding the ovary. The intensity of immunoreactivity for α-actin varied, with the cells of the vasculature reacting more intensely than the luteal cells. A difference in intensity of immunoreactivity was also observed among the luteal cells, with the inner granulosa cells showing stronger immunoreactivity than the peripheral theca lutein cells. There was no detectable immunoreactivity in the steroidogenic cells of the atretic corpus luteum. However, in both the active and atretic corpora lutea, α-actin-positive vascular cells were dispersed within the tissue. (b) Total α-actin (luteal and non-luteal), as determined by Western blot analyses, does not change during the luteal phase and subsequent corpus luteum demise (atretic corpora lutea). (c) hCG stimulated the expression of α-actin and progesterone secretion by the early luteal phase (LH surge + 1–5 days) and midluteal phase (LH surge + 6–10 days) cells in culture, but only progesterone in the late luteal phase (LH surge + 11–15 days). The data show that α-actin is present in luteal cells and that its expression is regulated by hCG, thus suggesting that E-cadherin may form functional adherens junctions in the corpus luteum.


2020 ◽  
Vol 12 (7) ◽  
pp. 543-555
Author(s):  
Yi Rang Na ◽  
Daun Jung ◽  
Juha Song ◽  
Jong-Wan Park ◽  
Jung Joo Hong ◽  
...  

Abstract Interleukin-10 (IL-10) is the most potent anti-inflammatory cytokine in the body and plays an essential role in determining outcomes of many inflammatory diseases. Cellular metabolism is a critical determinant of immune cell function; however, it is currently unclear whether metabolic processes are specifically involved in IL-10 production. In this study, we aimed to find the central metabolic molecule regulating IL-10 production of macrophages, which are the main producers of IL-10. Transcriptomic analysis identified that metabolic changes were predominantly enriched in Kupffer cells at the early inflammatory phase of a mouse endotoxemia model. Among them, pyruvate dehydrogenase kinase (PDK)-dependent acute glycolysis was negatively involved in IL-10 production. Inhibition or knockdown of PDK selectively increased macrophage IL-10 expression. Mechanistically, PDK inhibition increased IL-10 production via profound phosphorylation of adenosine monophosphate (AMP)-activated protein kinase alpha 1 (AMPKα1) by restricting glucose uptake in lipopolysaccharide-stimulated macrophages. AMPKα1 consequently activated p38 mitogen-activated protein kinase, c-Jun N-terminal kinase, and cyclic AMP-responsive element-binding protein to regulate IL-10 production. Our study uncovers a previously unknown regulatory mechanism of IL-10 in activated macrophages involving an immunometabolic function of PDK.


Sign in / Sign up

Export Citation Format

Share Document