Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 518
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Biol Reprod ; 111(3): 600-612, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-38910516

ABSTRACT

In cattle, the endometrium during diestrus and early pregnancy displays cellular responses that are consequences of prior, transient stimuli. Goal was to establish a model to study cellular memory in the endometrium. The hypothesis is that stimuli given to endometrium in vivo are retained as a cellular memory that remains after bovine uterine epithelial cells (BUECs) are isolated, cultured, and further stimulated in vitro. Objectives were to measure BUEC proliferation/migration and responsiveness to recombinant bovine Interferon-tau (rbIFNT) in vitro: among cows that showed estrus (experiment 1 [Exp1]), cows that became or not pregnant to artificial insemination (Exp2), cows that received or not supplemental progesterone (P4; Exp3) and cows that received or not a COX-1/2 inhibitor (Exp4). Only cows that displayed estrus were included in studies. For all experiments endometrial cytology was collected 4 days after estrus, BUECs were cultured, propagated, and submitted to rbIFNT treatment and an in vitro scratch assay. In Exp1, different cows spontaneously grouped according to proliferative/migratory capacity and responsiveness to rbIFNT of their respective BUECs. In Exp2, BUECs from pregnant cows showed greater rbIFNT responsiveness and cellular proliferation. In Exp3, BUECs from cows supplemented with P4 presented inhibited proliferation and increased expression of RSAD2. In Exp4, Flunixin Meglumine modified rbIFNT responsiveness of BUECs in an IFN-signaling pathway-specific manner. In conclusion, physiological and pharmacological stimuli received by the endometrium in vivo were retained as cellular memory in BUECs, persisted in culture, and changed BUEC proliferation/migration and responsiveness to rbIFNT, which are characteristics associated with fertility in cattle.


Subject(s)
Endometrium , Epithelial Cells , Interferon Type I , Uterus , Female , Animals , Cattle , Epithelial Cells/drug effects , Epithelial Cells/physiology , Interferon Type I/metabolism , Interferon Type I/pharmacology , Uterus/physiology , Uterus/drug effects , Endometrium/cytology , Endometrium/drug effects , Pregnancy , Cell Proliferation/drug effects , Pregnancy Proteins/pharmacology , Pregnancy Proteins/metabolism , Pregnancy Proteins/genetics , Cell Movement/drug effects , Progesterone/pharmacology , Cells, Cultured
2.
J Dairy Sci ; 107(9): 7371-7391, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38642656

ABSTRACT

Our objectives were to evaluate the endometrial responsiveness of dairy heifers to an intrauterine infusion of recombinant bovine interferon-tau (rbIFN-τ) and to associate endometrial responses to rbIFN-τ with subsequent reproductive performance. In experiments 1 and 2, cyclic heifers were enrolled in a program for synchronization of the estrous cycle, and blood sampling and ultrasonography examinations were performed on d 0, 4, 7, 11, and 14 of the estrous cycle. In experiment 1, heifers were randomly assigned to receive an intrauterine infusion containing 2 µg of rbIFN-τ (rbIFN-τ = 19) or saline control (CTRL = 19) into the uterine horn ipsilateral to the corpus luteum (CL) on d 14 of the estrous cycle. Then, 6 hours after the infusion, the infused uterine horn was flushed for sampling of the uterine luminal fluid (ULF) for composition analysis, and the endometrium was biopsied for transcriptomics. In experiment 2, 100 heifers received an intrauterine infusion of rbIFN-τ, and the same procedures for uterine sample collection were performed as described in experiment 1. After the intrauterine test, heifers were enrolled in a breeding program and classified as highly fertile (HF; pregnant at first AI) or subfertile (SF; not pregnant at first AI). Statistical analyses were performed using regression models, which included the effects of treatment (experiment 1: CTRL vs. rbIFN-τ) or fertility group (experiment 2: HF vs. SF) and block of samples. Intrauterine infusion of rbIFN-τ increased the expression of classical interferon-stimulated genes in the endometrium (e.g., ISG15, MX1, OAS2, IRF9, and USP18), and an antiviral response was predicted to be the main downstream effect of the transcriptome changes. In addition, rbIFN-τ increased the abundance of cholesterol, glycerol, and the overall concentration of oxylipins in the ULF. Analysis of endometrial transcriptome between HF and SF heifers revealed important differences in the expression of genes associated with cell signaling, metabolism, attachment, and migration, with a large representation of genes encoding extracellular matrix proteins. In general, differentially expressed genes were expected to be downregulated by IFN-τ but seemed to fail to be downregulated in SF heifers, resulting in higher expression in SF compared with HF heifers. Subfertile heifers had lower concentrations of glycerol and an altered profile of oxylipins in the ULF, with a lower abundance of oxylipins derived from arachidonic acid and dihomo-γ-linolenic acid, and a greater abundance of oxylipins derived from linoleic acid. Measurements of ovarian function did not differ between groups and, therefore, did not influence the observed results in uterine biology. Overall, the endometrial responsiveness to IFN-τ is variable among individuals and associated with subsequent fertility of heifers, indicating that communication between conceptus and endometrium is critical for the uterine receptivity and survival of pregnancy.


Subject(s)
Endometrium , Animals , Cattle , Female , Endometrium/drug effects , Endometrium/metabolism , Interferon Type I/pharmacology , Pregnancy , Reproduction/drug effects , Estrous Cycle/drug effects , Pregnancy Proteins/pharmacology
3.
Int J Mol Sci ; 25(14)2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39062763

ABSTRACT

Placental protein 13 (PP13) exhibits a plasma concentration that increases gradually during normal gestation, a process that is disrupted in preeclampsia, which is characterized by elevated vascular resistance, reduced utero-placental blood flow, and intrauterine growth restriction. This study investigated PP13's role in vascular tone regulation and its molecular mechanisms. Uterine and subcutaneous arteries, isolated from both pregnant and non-pregnant women, were precontracted with the thromboxane analogue U46619 and exposed to PP13 using pressurized myography. The molecular mechanisms were further investigated, using specific inhibitors for nitric oxide synthase (L-NAME+LNNA at 10-4 M) and guanylate cyclase (ODQ at 10-5 M). The results showed that PP13 induced vasodilation in uterine arteries, but not in subcutaneous arteries. Additionally, PP13 counteracted U46619-induced vasoconstriction, which is particularly pronounced in pregnancy. Further investigation revealed that PP13's mechanism of action is dependent on the activation of the nitric oxide-cGMP pathway. This study provides novel insights into the vasomodulatory effects of PP13 on human uterine arteries, underscoring its potential role in regulating utero-placental blood flow. These findings suggest that PP13 may be a promising candidate for improving utero-placental blood flow in conditions such as preeclampsia. Further research and clinical studies are warranted to validate PP13's efficacy and safety as a therapeutic agent for managing preeclampsia.


Subject(s)
Pre-Eclampsia , Pregnancy Proteins , Uterine Artery , Humans , Female , Pre-Eclampsia/metabolism , Pregnancy , Uterine Artery/metabolism , Uterine Artery/drug effects , Adult , Pregnancy Proteins/metabolism , Pregnancy Proteins/pharmacology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Vasodilation/drug effects , Nitric Oxide/metabolism , Vasoconstriction/drug effects , Cyclic GMP/metabolism , Placenta/metabolism , Placenta/blood supply , Placenta/drug effects , Galectins
4.
Biochem Biophys Res Commun ; 553: 37-43, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33765557

ABSTRACT

Previously, we reported that the presence of multiple day 7 (D7) bovine embryos in the uterus induces systemic immune responses in circulating polymorphonuclear neutrophils (PMNs), but with unknown mechanism. Thus, this study aimed to investigate the direct impact of D7 bovine embryo on PMNs' immune responses in vitro and whether these PMNs can amplify and transfer embryo signals further to another PMN population. PMNs were directly stimulated by embryo culture media (ECM) or interferon tau (IFNT) (10 ng/ml) followed by evaluating mRNA expression by real-time PCR and phenotypic analysis by flow cytometry. To test whether PMNs can transfer embryo signals to a new PMN population, PMNs triggered by ECM or IFNT, were thoroughly washed and diluted to remove any media components, and again were incubated in fresh culture media for 3 h, from which culture supernatants were collected and used as PMN conditioned media (CM) to stimulate a new PMN population. Similar to ECM, IFNT directly stimulated expressions of IFNs (IFNA, IFNG), interferon-stimulated genes (ISGs; OAS1, ISG15, MX1), STAT1, TGFB and IL8, and downregulated TNFA in PMNs. Flow cytometrical analyses demonstrated that IFNT stimulated expressions of pregnancy-related phenotypic markers, CD16 and arginase-1 (ARG1), in PMNs. Most importantly, PMN CM induced ISGs and STAT1 mRNA in fresh PMNs. Since IFNT directly upregulated IFNA expression in PMNs, the impact of IFNA on PMNs' immune responses was further tested. Stimulation of PMNs with IFNA, especially at a low level (1 pg/ml), induced IFNT-like immune responses comparable to those induced by PMN CM. Together, these findings indicated that D7 bovine embryos induce direct anti-inflammatory responses with upregulation of ISGs expressions in PMNs mainly via IFNT. Additionally, PMNs can amplify and transfer embryo signals to a new PMN population in a cell-to-cell communication mechanism possibly mediated in part by IFNA. Such a novel immunological crosstalk might contribute to embryo tolerance and pregnancy establishment in cattle.


Subject(s)
Embryo, Mammalian/immunology , Embryo, Mammalian/metabolism , Gene Expression Regulation , Interferon Type I/immunology , Neutrophils/immunology , Pregnancy Proteins/immunology , Pregnancy/genetics , Pregnancy/immunology , Animals , Arginase/genetics , Cattle , Culture Media, Conditioned/pharmacology , Female , Gene Expression Regulation/drug effects , Immunity, Innate , In Vitro Techniques , Interferon Type I/pharmacology , Neutrophils/drug effects , Neutrophils/metabolism , Phenotype , Pregnancy Proteins/pharmacology , Receptors, IgG/genetics
5.
Reprod Fertil Dev ; 33(4): 305-317, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33573713

ABSTRACT

In cattle, maternal recognition of early pregnancy depends on the effects of the embryonic signal interferon (IFN)-τ. IFN-stimulated genes have been upregulated in the maternal liver during early pregnancy. In this study, primary hepatocyte cell culture models were evaluated for their suitability to test Type I IFN effects invitro. The expression of target genes (interferon-stimulated gene 15 (ISG-15), interferon-induced GTP-binding protein (MX-1), C-X-C motif chemokine 10 (CXCL-10), CXCL-5, insulin-like growth factor 1 (IGF-1), IGF binding protein 2 (IGFBP-2)) was measured using reverse transcription-quantitative polymerase chain reaction in hepatocytes from monoculture or in indirect coculture with Kupffer cells (HKCid) on Days 1, 2, 3 and 4 of culture (n=21 donor cows). Gene expression was also measured on Day 4 after challenging the cultures with recombinant IFNτ, IFNα, progesterone (P4), IFNτ+IFNα or IFNτ+P4 for 6h. A significant increase in the mRNA expression of target genes in hepatocytes was shown in response to stimulation with IFNτ. The Kupffer cells in coculture did not influence the effects of IFNτ in hepatocytes. In conclusion, primary bovine hepatocyte cultures are suitable for stimulation experiments with Type I IFNs and as an extrauterine model for embryo-maternal communication. The proposed endocrine action of IFNτ in the liver may affect maternal metabolism and immune function in the liver.


Subject(s)
Hepatocytes/drug effects , Interferon Type I/pharmacology , Kupffer Cells/metabolism , Liver/drug effects , Animals , Cattle , Cell Communication , Cells, Cultured , Coculture Techniques , Gene Expression Regulation , Hepatocytes/immunology , Hepatocytes/metabolism , Interferon-alpha/pharmacology , Liver/immunology , Liver/metabolism , Pregnancy Proteins/pharmacology , Progesterone/pharmacology
6.
Int J Mol Sci ; 22(10)2021 May 18.
Article in English | MEDLINE | ID: mdl-34070207

ABSTRACT

In domestic ruminants, endometrial receptivity is related to successful pregnancy and economic efficiency. Despite several molecules having been reported in the past regarding endometrial receptivity regulation, much regarding the mechanism of endometrial receptivity regulation remains unknown due to the complex nature of the trait. In this work, we demonstrated that the cysteine-rich transmembrane bone morphogenetic protein (BMP) regulator 1 (CRIM1) served as a novel regulator in the regulation of goat endometrial receptivity in vitro. Our results showed that hormones and IFN-τ increased the expression of CRIM1 in goat endometrial epithelial cells (EECs). Knockdown of CRIM1 via specific shRNA hindered cell proliferation, cell adhesion and prostaglandins (PGs) secretion and thus derailed normal endometrial receptivity. We further confirmed that receptivity defect phenotypes due to CRIM1 interference were restored by ATG7 overexpression in EECs while a loss of ATG7 further impaired receptivity phenotypes. Moreover, our results showed that changing the expression of ATG7 affected the reactive oxygen species (ROS) production. Moreover, mR-143-5p was shown to be a potential upstream factor of CRIM1-regulated endometrial receptivity in EECs. Overall, these results suggest that CRIM1, as the downstream target of miR-143-5p, has effects on ATG7-dependent autophagy, regulating cell proliferation, cell adhesion and PG secretion, and provides a new target for the diagnosis and treatment of early pregnancy failure and for improving the success rates of artificial reproduction.


Subject(s)
Bone Morphogenetic Protein Receptors/physiology , Embryo Implantation/genetics , Endometrium/physiology , Goats/physiology , Animals , Autophagy/drug effects , Autophagy/genetics , Autophagy/physiology , Autophagy-Related Protein 7/deficiency , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/physiology , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Protein Receptors/genetics , Cell Adhesion , Cell Proliferation , Cells, Cultured , Embryo Implantation/physiology , Endometrium/cytology , Endometrium/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/physiology , Estradiol/pharmacology , Female , Gene Knockdown Techniques , Goats/genetics , Interferon Type I/pharmacology , MicroRNAs/genetics , MicroRNAs/metabolism , Models, Biological , Pregnancy , Pregnancy Proteins/pharmacology , Progesterone/pharmacology , Prostaglandins/metabolism , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Up-Regulation
7.
Biochem Biophys Res Commun ; 532(1): 101-107, 2020 10 29.
Article in English | MEDLINE | ID: mdl-32828539

ABSTRACT

Uterine infection with bacteria and the release of peptidoglycan (PGN), antigenic cell wall components of both Gram-negative and Gram-positive bacteria, can cause early pregnancy losses in ruminants, but the associated mechanisms remain unsolved. Day 7 blastocyst starts to secrete a minute amount of interferon-tau (IFNT) in the uterine horn which is required for early stage of maternal recognition of pregnancy (MRP) in ruminants, and it induces interferon stimulated genes (ISGs) for driving uterine receptivity in cows. This study investigated if PGN disrupts IFNT response through modulation of endometrial ISGs expressions. Cultured bovine endometrial epithelial cells (BEECs) were treated with embryo culture medium (ECM) or IFNT (1 ng/ml) in the presence or absence of a low level of PGN (10 pg/ml) for 24 h. A real-time PCR analyses revealed that the presence of PGN suppressed IFNT-induced ISGs (OAS1 and ISG15) and STAT1 expressions in BEECs. To visualize the impact of PGN in an ex-vivo model that resembles the in vivo status, endometrial explants were treated by IFNT (1 ng/ml) with or without PGN (10 pg/ml) for 12 h. PGN suppressed IFNT-induced gene expressions of the above factors, but not for IFNA receptor type1 (IFNAR1) or type2 (IFNAR2) in explants. Immunofluorescence analysis illustrated that PGN completely suppressed the IFNT-triggered OAS1 protein expression in the luminal epithelium of explants. Of note, PGN did not stimulate pro-inflammatory cytokines (TNFA and IL1B) or TLR2 mRNA expression in both models. These findings indicate that the presence of low levels of PGN suppresses ISGs expression induced by IFNT secreted from early embryo, at the luminal epithelium of the bovine endometrium. This could severely interfere with early stage of MRP processes in cows, leading to pregnancy failure.


Subject(s)
Endometrium/metabolism , Interferon Type I/metabolism , Peptidoglycan/metabolism , Pregnancy Proteins/metabolism , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , Abortion, Veterinary/immunology , Abortion, Veterinary/metabolism , Abortion, Veterinary/microbiology , Animals , Blastocyst/immunology , Blastocyst/metabolism , Blastocyst/microbiology , Cattle , Cattle Diseases/genetics , Cattle Diseases/metabolism , Cattle Diseases/microbiology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Endometrium/immunology , Endometrium/microbiology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Female , Gene Expression , In Vitro Techniques , Interferon Type I/pharmacology , Maternal-Fetal Exchange/immunology , Peptidoglycan/immunology , Pregnancy , Pregnancy Proteins/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT1 Transcription Factor/genetics , Uterine Diseases/genetics , Uterine Diseases/metabolism , Uterine Diseases/veterinary , Uterus/immunology , Uterus/metabolism , Uterus/microbiology
8.
Reproduction ; 160(4): 603-612, 2020 10.
Article in English | MEDLINE | ID: mdl-32698154

ABSTRACT

Pentraxin 3 (PTX3), a multimeric glycoprotein, is implicated in various biological functions. PTX3 was shown to be elevated in the corpus luteum (CL) of early pregnant ewes; however, its role in sheep or other ruminants' CL during this reproductive stage or how it is regulated remain unknown. Here we explored the role of PTX3 and its relationship with interferon-tau (IFNT; the pregnancy recognition signaling molecule during early pregnancy in domestic ruminants) in bovine luteinized granulosa cells (LGCs). IFNT robustly elevated PTX3 expression in bovine LGCs, and significantly stimulated its expression in luteal endothelial cells, along with CL slices; yet, LGCs were the most responsive and sensitive among these luteal models. ALK2/ALK3/ALK6 kinase inhibitor, dorsomorphin, dose-dependently inhibited basal and IFNT-elevated PTX3 expression in LGCs. In contrast, ALK4/5/7 inhibitor, SB431542, did not alter basal and TGFB1-induced PTX3. We found that recombinant human PTX3 itself moderately but significantly increases LGC numbers. Because PTX3 is highly expressed in bovine LGCs, we next examined the impact of lowering endogenous PTX3 levels with siRNA. PTX3 silencing decreased the viable cell numbers and reversed IFNT actions on cell viability, percentage of proliferating cells, and on two key survival/death genes: BIRC5 encoding surviving protein, and FASL - a death-inducing signal. Interestingly, thrombospondin-1, a known luteal proapoptotic factor, was inversely related to PTX3 in LGCs. Together, these findings suggest a novel role for PTX3 during early pregnancy, as mediator of IFNT prosurvival actions supporting CL maintenance during this reproductive stage.


Subject(s)
C-Reactive Protein/metabolism , Corpus Luteum/cytology , Gene Expression Regulation/drug effects , Interferon Type I/pharmacology , Luteal Cells/cytology , Pregnancy Proteins/pharmacology , Serum Amyloid P-Component/metabolism , Animals , C-Reactive Protein/genetics , Cattle , Corpus Luteum/drug effects , Corpus Luteum/metabolism , Female , Luteal Cells/drug effects , Luteal Cells/metabolism , Pregnancy , Serum Amyloid P-Component/genetics
9.
J Cell Physiol ; 234(4): 4864-4873, 2019 04.
Article in English | MEDLINE | ID: mdl-30238980

ABSTRACT

Ephrin A1 has a role in a variety of biological events, including cell proliferation, differentiation, migration, and angiogenesis. Ephrin A1 expression is abundant in trophoblasts and endometrial cells during the implantation period; however, its intracellular activities have not yet been reported in bovine endometrial (BEND) epithelial cells. The aim of this study was to identify the functional role of ephrin A1 in BEND cells, which have served as a good model system for investigating the regulation of signal transduction following treatment with interferon-τ (IFNT) in vitro. Supplementation of ephrin A1 to BEND cells increased cell proliferation and increased levels of proliferating cell nuclear antigen and cyclin D1 protein in BEND cell nuclei. To investigate intracellular mechanisms regulated by ephrin A1, we performed Western blot analysis focused on mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) signaling, which are significantly involved in the successful maintenance of pregnancy. Ephrin A1 dose-dependently increased phosphorylation of extracellular signal-regulated kinases (ERK)1/2, c-Jun N-terminal kinases (JNK), P38, protein kinase B (AKT), P70S6K, S6, and cyclin D1, and the activated proteins were suppressed by pharmacological inhibitors including wortmannin (a PI3K inhibitor), U0126 (an ERK1/2 inhibitor), and SP600125 (a JNK inhibitor). Among ephrin A1 receptors, abundant expression of EPHA2 and EPHA4 messenger RNA was detected in BEND cells by reverse transcription polymerase chain reaction analysis. Furthermore, tunicamycin-induced endoplasmic reticulum (ER) stress was inactivated by ephrin A1 treatment of BEND cells. Our findings suggest that ephrin A1 promotes the development of BEND cells and likely enhances uterine capacity and maintenance of pregnancy by activating MAPK and PI3K signaling cascades and by restoring ER stress.


Subject(s)
Cyclin D1/metabolism , Endometrium/metabolism , Ephrin-A1/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Animals , Anthracenes/pharmacology , Butadienes/pharmacology , Cattle , Cell Cycle/physiology , Cell Line , Cell Proliferation , Endometrium/cytology , Endoplasmic Reticulum Stress/physiology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Interferon Type I/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Nitriles/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Pregnancy , Pregnancy Proteins/pharmacology , Receptor, EphA2/metabolism , Receptor, EphA4/metabolism , Wortmannin/pharmacology
10.
Gynecol Endocrinol ; 35(sup1): 35-40, 2019.
Article in English | MEDLINE | ID: mdl-31532312

ABSTRACT

Preeclampsia still remains one of the most severe pregnancy complications and is an actual problem in the obstetrics practice. At present, the joint impact of cytokines and other placenta secreted factors on trophoblast cell functional activity during preeclampsia complicated pregnancy remains unclear. The aim of the study is to estimate the surface receptors expression by trophoblast cells in the presence of placenta secreted factors during physiological pregnancy and at preeclampsia. Trophoblast cells of the JEG-3 line were incubated in the presence of supernatants obtained by cultivation of placentas from women with physiological pregnancy and with preeclampsia. Surface receptors expression by trophoblast cells was estimated by FACS Canto II flow cytometer. It was established that in the third trimester both under normal and pathological conditions, the placenta secreted factors impact on the cytokine receptor expression by trophoblast differs while the trophoblast response capacity to the migration and proliferation stimulating and inhibiting signals remains stable. JEG-3 line cells enhanced the expression of CD186, CD140a, Integrin ß6, VE-cadherin, CD29, and CD140a in the case of incubation in the presence of placenta supernatants from the third-trimester pregnancy complicated with preeclampsia compared to incubation in the presence of placenta supernatants form the third trimester of physiological pregnancy.


Subject(s)
Pregnancy Proteins/pharmacology , Receptors, Cell Surface/genetics , Trophoblasts/drug effects , Trophoblasts/metabolism , Adult , Case-Control Studies , Cells, Cultured , Cytokines/metabolism , Cytokines/pharmacology , Female , Gene Expression/drug effects , Humans , Placenta/metabolism , Placenta/pathology , Placental Hormones/metabolism , Placental Hormones/pharmacology , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Pregnancy , Pregnancy Proteins/metabolism , Receptors, Cell Surface/metabolism
11.
J Cell Physiol ; 233(6): 4885-4894, 2018 06.
Article in English | MEDLINE | ID: mdl-29194593

ABSTRACT

Interferon tau, a 23 kDa trophoblast derived protein diffuses out from the uterus into the circulation and leads to the expression of IFNτ stimulated genes viz. ISG15 and OAS1 in blood neutrophils. The IFNτ pathway is species as well as tissue specific. To unsnarl the IFNτ downstream signaling pathway, the blood neutrophils were incubated simultaneously with 10 ng/ml of recombinant bovine interferon tau and the inhibitors of JAK2 (AG490), JAK3 (CP690550), p38 (SB202190), PI3K/Akt (LY294002), and MAPK/Erk (U0126) at specific doses for 4-hr duration. The IFNτ pathway was determined through real-time gene expression of ISG15 and OAS1; immunocytochemistry of ISG15; and Western blotting of ISG15, OAS1, pJAK3 and PI3K. The ISG15 and OAS1 expression decreased significantly (p < 0.001) in the presence of pJAK3 and PI3K inhibitors as compared to a positive control where only interferon tau was used. Immunocytochemistry revealed an attenuated ISG15 response while stimulating blood neutrophils with pJAK3 inhibitor (CP690550) and PI3K inhibitor (LY294002). Similarly, Western blot analysis of neutrophil protein fraction showed weak signals of ISG15, OAS1, pJAK3 and PI3K in the presence of pJAK3 and PI3K inhibitors. The expression profile, immunocytochemistry and western blot analysis revealed a JAK3 and PI3K mediated interferon-tau stimulated gene expression in blood neutrophils.


Subject(s)
Gene Expression Regulation/drug effects , Interferon Type I/pharmacology , Janus Kinase 3/metabolism , Neutrophils/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Pregnancy Proteins/pharmacology , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , Animals , Cattle , Cells, Cultured , Female , Neutrophil Activation/drug effects , Neutrophils/enzymology , Phagocytosis/drug effects , Phosphorylation , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Ubiquitins/genetics , Ubiquitins/metabolism
12.
J Reprod Dev ; 64(6): 495-502, 2018 Dec 14.
Article in English | MEDLINE | ID: mdl-30298824

ABSTRACT

Interferon-tau (IFNT), a type I interferon (IFN), is known as pregnancy recognition signaling molecule secreted from the ruminant conceptus during the preimplantation period. Type I IFNs, such as IFN-alpha and IFN-beta, are known to activate cell-death pathways as well as induce apoptosis. In cows, induction of apoptosis with DNA fragmentation is induced by IFNT in cultured bovine endometrial epithelial cells. However, the status of cell-death pathways in the bovine endometrium during the preimplantation period still remains unclear. In the present study, we investigated the different cell-death pathways, including apoptosis, pyroptosis, and autophagy, in uterine tissue obtained from pregnant cows and in vitro cultured endometrial epithelial cells with IFNT stimulation. The expression of CASP7, 8, and FADD (apoptosis-related genes) was significantly higher in pregnant day 18 uterine tissue in comparison to non-pregnant day 18 tissue. The expression of CASP4, 11, and NLRP3 (pyroptosis-related genes) was significantly higher in the pregnant uterus in comparison to non-pregnant uterus. In contrast, autophagy-related genes were not affected by pregnancy. We also investigated the effect of IFNT on the expression of cell-death pathway-related genes, as well as DNA fragmentation in cultured endometrial epithelial cells. Similar to its effects in pregnant uterine tissue, IFNT affected the increase of apoptosis-related (CASP8) and pyroptosis-related genes (CASP11), but did not affect autophagy-related gene expression. IFNT also increased γH2AX-positive cells, which is a marker of DNA fragmentation. These results suggest that apoptosis- and pyroptosis-related genes are induced by IFNT in the pregnant bovine endometrial epithelial cells.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Endometrium/drug effects , Interferon Type I/pharmacology , Pregnancy Proteins/pharmacology , Pyroptosis/drug effects , Signal Transduction/drug effects , Animals , Caspase 7/metabolism , Caspase 8/metabolism , Cattle , DNA Fragmentation/drug effects , Endometrium/metabolism , Female , Pregnancy
13.
Cell Physiol Biochem ; 43(6): 2277-2296, 2017.
Article in English | MEDLINE | ID: mdl-29073617

ABSTRACT

BACKGROUND/AIMS: Pregnancy success requires mandatory maternal tolerance of the semi/ allogeneic embryo involving embryo-derived signals. Expression levels of PreImplantation Factor (PIF), a novel peptide secreted by viable embryos, correlate with embryo development, and its early detection in circulation correlates with a favourable pregnancy outcome. PIF enhances endometrial receptivity to promote embryo implantation. Via the p53 pathway, it increases trophoblast invasion, improving cell survival / immune privilege. PIF also reduces spontaneous and LPS-induced foetal death in immune naïve murine model. We examined PIF effect on gene expression of human leukocyte antigen (HLA-G, -E -F and -C) and the influence of PIF on local progesterone activity in JEG-3 choriocarcinoma cells. METHODS: PIF and progesterone (P4) effects on JEG-3 cells surface and intracellular HLA molecules was tested using monoclonal antibodies, flow cytometry, and Western blotting. PIF and IL17 effects on P4 and cytokines secretion was determined by ELISA. PIF and P4 effects on JEG-3 cells proteome was examined using 2D gel staining followed by spot analysis, mass spectrometry and bioinformatic analysis. RESULTS: In cytotrophoblastic JEG-3 cells PIF increased intracellular expression of HLA-G, HLA-F, HLA-E and HLA-C and surface expression of HLA-G, HLA-E and HLA-C in dose and time dependent manner. In case of HLA-E, -F results were confirmed also by Western blot. Proteome analysis confirmed an increase in HLA-G, pro-tolerance FOXP3+ regulatory T cells (Tregs), coagulation factors and complement regulator. In contrast, PIF reduced PRDX2 and HSP70s to negate oxidative stress and protein misfolding. PIF enhanced local progesterone activity, increasing steroid secretion and the receptor protein. It also promoted the secretion of the Th1/Th2 cytokines (IL-10, IL-1ß, IL-8, GM-CSF and TGF-ß1), resulting in improved maternal signalling. CONCLUSION: PIF can generate a pro-tolerance milieu by enhancing the expression of HLA molecules and by amplifying endogenous progesterone activity. A Fast-Track clinical trial for autoimmune disease has been satisfactorily completed. The acquired data warrants PIF use for the treatment of early pregnancy disorders.


Subject(s)
Gene Expression/drug effects , HLA-C Antigens/metabolism , HLA-G Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Pregnancy Proteins/pharmacology , Progesterone/pharmacology , Cell Line, Tumor , Choriocarcinoma/metabolism , Choriocarcinoma/pathology , Cluster Analysis , Cytokines/analysis , Electrophoresis, Gel, Two-Dimensional , Enzyme-Linked Immunosorbent Assay , Female , HLA-C Antigens/genetics , HLA-G Antigens/genetics , HSP70 Heat-Shock Proteins/metabolism , Histocompatibility Antigens Class I/genetics , Humans , Interleukin-17/pharmacology , Peptides/analysis , Peroxiredoxins/metabolism , Pregnancy , Pregnancy Proteins/genetics , Pregnancy Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology , HLA-E Antigens
14.
Reproduction ; 154(5): F45-F59, 2017 11.
Article in English | MEDLINE | ID: mdl-28982937

ABSTRACT

This review focuses on the paracrine and endocrine actions of interferon tau (IFNT) during pregnancy recognition and establishment in ruminants. Pregnancy recognition involves the suppression of the endometrial luteolytic mechanism by the conceptus to maintain progesterone production by the corpus luteum (CL). The paracrine antiluteolytic effects of conceptus-derived IFNT inhibit upregulation of oxytocin receptors in the endometrial epithelia of the uterus, thereby preventing the production of luteolytic prostaglandin F2 alpha (PGF2α) pulses. In the endometrium, IFNT induces or upregulates a large number of classical IFN-stimulated genes (ISGs) and regulates expression of many other genes in a cell-specific manner that are likely important for conceptus elongation, implantation and establishment of pregnancy. Further, IFNT has endocrine effects on extrauterine cells and tissues. In sheep, IFNT induces luteal resistance to PGF2α, thereby ensuring survival of the CL for maintenance of pregnancy. The ISGs induced in circulating peripheral blood mononuclear cells by IFNT may also be useful as an indicator of pregnancy status in cattle. An increased knowledge of IFNT and ISGs is important to improve the reproductive efficiency in ruminants.


Subject(s)
Interferon Type I/pharmacology , Interferon Type I/physiology , Paracrine Communication/drug effects , Pregnancy Proteins/pharmacology , Pregnancy Proteins/physiology , Animals , Embryo Implantation/drug effects , Endocrine System/drug effects , Female , Humans , Luteolysis/drug effects , Paracrine Communication/physiology , Pregnancy , Ruminants
15.
Reproduction ; 154(5): 559-568, 2017 11.
Article in English | MEDLINE | ID: mdl-28808111

ABSTRACT

Interferon-tau (IFNT), a maternal recognition of pregnancy (MRP) signals in domestic ruminants, suppresses the release of luteolytic pulses of uterine prostaglandin F2a (PGF2a), thus extending the corpus luteum (CL) life span. We hypothesized that IFNT also exerts anti-luteolytic actions in bovine CL. To examine the direct effects of IFNT on bovine CL, luteal slices and enriched luteal endothelial cells (LECs) were utilized. We found that recombinant ovine IFNT (roIFNT) markedly elevates interferon-associated genes (STAT1, STAT2 and IRF9) and interferon-stimulated genes (ISGs: MX2, ISG15 and OAS1Y) in both models. Furthermore, IFNT time-dependently induced STAT1 phosphorylation in LECs without affecting total STAT1. roIFNT-stimulated viable LECs numbers and the knockdown of protein inhibitor of activated STAT1 (PIAS1) abolished this effect, suggesting that PIAS1 may mediate the proliferative effect of IFNT. IFNT significantly downregulated luteolytic genes such as TGFB1, thrombospondin-1 (THBS1), endothelin-1 (EDN1) and serpin family E member-1 (SERPINE1) in LECs. However, less robust effects were observed in luteal slices. Moreover, PGF2a alone induced THBS1, SERPINE1 and EDN1 mRNA in CL slices whereas in the presence of IFNT, THBS1 and SERPINE1 stimulation was abolished. Collectively, these results indicate that IFNT acts via STAT1- IRF9-dependent and independent pathways and affects diverse luteal functions. Most interestingly, this study suggests the existence of an anti-luteolytic effect of IFNT in bovine CL, namely, inhibiting key PGF2a-induced luteolytic genes. The proliferative effect of IFNT may constitute an additional mechanism that promotes luteal cell survival, thus, extending the luteal life span during early pregnancy in cows.


Subject(s)
Cattle , Corpus Luteum/drug effects , Endothelial Cells/drug effects , Interferon Type I/pharmacology , Luteolysis/drug effects , Luteolysis/genetics , Pregnancy Proteins/pharmacology , Pregnancy, Animal , Animals , Cattle/genetics , Cattle/metabolism , Cell Survival/drug effects , Cells, Cultured , Corpus Luteum/cytology , Corpus Luteum/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Luteal Cells/drug effects , Pregnancy
16.
J Immunol ; 194(6): 2513-21, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25694608

ABSTRACT

Inflammation-mediated oncogenesis has been implicated in a variety of cancer types. Rheumatoid synovial tissues can be viewed as a tumor-like mass, consisting of hyperplastic fibroblast-like synoviocytes (FLSs). FLSs of rheumatoid arthritis (RA) patients have promigratory and invasive characteristics, which may be caused by chronic exposure to genotoxic stimuli, including hypoxia and growth factors. We tested whether a transformed phenotype of RA-FLSs is associated with placental growth factor (PlGF), a representative angiogenic growth factor induced by hypoxia. In this study, we identified PlGF-1 and PlGF-2 as the major PlGF isoforms in RA-FLSs. Global gene expression profiling revealed that cell proliferation, apoptosis, angiogenesis, and cell migration were mainly represented by differentially expressed genes in RA-FLSs transfected with small interfering RNA for PlGF. Indeed, PlGF-deficient RA-FLSs showed a decrease in cell proliferation, migration, and invasion, but an increase in apoptotic death in vitro. PlGF gene overexpression resulted in the opposite effects. Moreover, exogeneous PlGF-1 and PlGF-2 increased survival, migration, and invasiveness of RA-FLSs by binding their receptors, Flt-1 and neuropilin-1, and upregulating the expression of antiapoptotic molecules, pErk and Bcl2. Knockdown of PlGF transcripts reduced RA-FLS proliferation in a xenotransplantation model. Collectively, in addition to their role for neovascularization, PlGF-1 and -2 promote proliferation, survival, migration, and invasion of RA-FLSs in an autocrine and paracrine manner. These results demonstrated how primary cells of mesenchymal origin acquired an aggressive and transformed phenotype. PlGF and its receptors thus offer new targets for anti-FLS therapy.


Subject(s)
Cell Movement/genetics , Cell Proliferation/genetics , Pregnancy Proteins/genetics , Synovial Membrane/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Arthritis, Rheumatoid/pathology , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Profiling , HEK293 Cells , Humans , Hyperplasia/genetics , Microscopy, Confocal , Neovascularization, Pathologic/genetics , Oligonucleotide Array Sequence Analysis , Placenta Growth Factor , Pregnancy Proteins/metabolism , Pregnancy Proteins/pharmacology , Primary Cell Culture , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Synovial Membrane/blood supply , Synovial Membrane/pathology , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
17.
Reprod Fertil Dev ; 29(6): 1209-1216, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27165775

ABSTRACT

Interferon (IFN)-stimulated gene 15 (ISG15) is one of several proteins induced by conceptus-derived Type I or II IFNs in the uterus, and is implicated as an important factor in determining uterine receptivity to embryos in ruminants. But little is known about the role the ISG15 gene or gene product plays during embryo development. In the present study, both the expression profile and function of ISG15 were investigated in early bovine embryos in vitro. ISG15 mRNA was detectable in Day 0, 2, 6 and 8 bovine embryos, but IFN-τ (IFNT) mRNA only appeared from Day 6. This means that embryonic expression of ISG15 on Days 0 and 2 was not induced by embryonic IFNT. However, ISG15 mRNA expression paralleled the expression of IFNT mRNA in Day 6 and 8 embryos. ISG15-lentivirus interference plasmid (ISG15i) was injected into 2-cell embryos to knockdown ISG15 expression. This resulted in decreases in the proportion of hatching blastocysts, the diameter of blastocysts and cell number per diameter of blastocysts compared with control embryos. In addition, ISG15i inhibited IFNT, Ets2 (E26 oncogene homolog 2) mRNA and connexion 43 protein expression in Day 8 blastocysts, whereas exogenous IFNT treatment (100ngmL-1, from Day 4 to Day 8) improved ISG15 mRNA and connexion 43 protein expression. In conclusion, it appears that ISG15 is involved in early bovine embryo development and that it regulates IFNT expression in the blastocyst.


Subject(s)
Blastocyst/metabolism , Cytokines/metabolism , Ectogenesis , Gene Expression Regulation, Developmental , Interferon Type I/metabolism , Pregnancy Proteins/metabolism , Ubiquitins/metabolism , Up-Regulation , Abattoirs , Animals , Animals, Inbred Strains , Blastocyst/cytology , Blastocyst/drug effects , Cattle , Cryopreservation , Cytokines/antagonists & inhibitors , Cytokines/genetics , Ectogenesis/drug effects , Female , Fertilization in Vitro , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Vitro Oocyte Maturation Techniques , Interferon Type I/genetics , Interferon Type I/pharmacology , Male , Pregnancy Proteins/genetics , Pregnancy Proteins/pharmacology , RNA Interference , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Semen Preservation , Ubiquitins/antagonists & inhibitors , Ubiquitins/genetics , Up-Regulation/drug effects
18.
Dokl Biol Sci ; 475(1): 180-182, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28861873

ABSTRACT

The effect of human pregnancy-specific glycoprotein (PSG) on the cytokine and chemokine production in vitro by intact mononuclear cells was studied by the method of flow fluorimetry. PSG inhibited production of the proinflammatory cytokines IL-6, IL-8, IL-17, IFN-γ, and TNF-α and chemokines CCL3/MIP-1α, CCL4/MIP-1ß, CCL2/MCP-1; at the same time, PSG stimulated IL-12(p70) production. Simultaneously with increasing the VEGF level, PSG inhibited production of IL-9, IL-13, G-CSF, and GM-CSF. The PSG effect discovered can be interpreted as a contribution into the immune tolerance formation during pregnancy.


Subject(s)
Cytokines/biosynthesis , Leukocytes, Mononuclear/metabolism , Pregnancy Proteins/pharmacology , Female , Humans , Pregnancy , Pregnancy Proteins/blood
19.
Reprod Biol Endocrinol ; 14: 22, 2016 Apr 18.
Article in English | MEDLINE | ID: mdl-27091464

ABSTRACT

BACKGROUND: In ruminants, embryo implantation depends on progesterone (P4) and interferon tau (IFNT) controlling endometrial function. IFNT antagonizes bovine endometrial cells (BEND) response to phorbol 12,13-dibutyrate (PDBU) through posttranscriptional regulation of gene expression. We have previously described microRNAs (miRNAs) profiles in bovine endometrium, detecting miR-106a, relevant for embryo maternal communication. In this study, we investigated the expression miR-106a and genes for prostaglandin-endoperoxide synthase 2 (PTGS2), phospholipase A2, group IVA (PLA2G4A), estrogen receptor 1 (ESR1) and progesterone receptor (PR) in response to IFNT in BEND cells and searched for interferon responsive factors (IRFs) binding sites in their promoter genomic regions. The aim of this study was to unravel the molecular mechanisms involved in IFNT signalling and its regulation of miR-106a. FINDINGS: PTGS2 showed increased expression under PDBU, which was antagonized by IFNT. IFNT induced expression of PR and miR-106a and downregulation of ESR1 and PR. Bioinformatic analyses detected that PLA2G4A was associated to IRF-1 and IRF-6, while ESR1, PR and PTGS2 were associated to only IRF-6. All genes exhibit one motif per IRF, except miR-106a that had three binding sites for IRF-6. CONCLUSIONS: We report the IFNT regulatory effect on miR-106a expression through IRF-6 in bovine endometrial cells. We identified a set of potential binding sites for IRF-1 and IRF-6 within the bovine genome. A set of candidate gene regions could be characterized where IFNT can act via IRFs to regulate the expression of proteins and miRNAs. Future studies will use these data to detect new IFNT regulatory mechanisms in the endometrium.


Subject(s)
Endometrium/cytology , Gene Expression Regulation, Developmental , Interferon Type I/pharmacology , Pregnancy Proteins/pharmacology , Animals , Binding Sites , Cattle , Cells, Cultured , Computational Biology , Cyclooxygenase 2/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Group IV Phospholipases A2/genetics , Group IV Phospholipases A2/metabolism , Interferon Type I/genetics , Interferon Type I/metabolism , MicroRNAs/metabolism , Pregnancy Proteins/genetics , Pregnancy Proteins/metabolism , Promoter Regions, Genetic , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Signal Transduction
20.
J Immunol ; 192(7): 3003-10, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24574497

ABSTRACT

Bone marrow-derived mesenchymal stem cells (MSC) exist in the synovium of patients with rheumatoid arthritis (RA), yet the role of MSC in RA is elusive. Placental growth factor (PlGF) expression is increased in RA synovial fluids, and blocking of PlGF attenuates progression of arthritis in mice. In this study, we observed that PlGF induced chemotaxis of MSC in a dose-dependent manner, which was blocked by anti-vascular endothelial growth factor receptor-1 peptide. MSC exposed to PlGF elicited increased phosphorylation of Akt and p38 MAPK. PlGF-mediated chemotaxis was inhibited by PI3K inhibitor (LY294002) and p38 MAPK inhibitor (SB203580), but not by ERK1/2 inhibitor (PD98059). Fibroblast-like synoviocytes (FLS) constitutively produced PlGF, but MSC released negligible amounts of PlGF. Of note, when FLS of RA patients and MSC were cocultured, PlGF production by FLS was significantly increased; such an increase was dependent on the number of added MSC. Moreover, coculture conditioned medium promoted chemotaxis of MSC and increased angiogenesis in Matrigel plugs assay, and these were suppressed by preincubation of the medium with anti-PlGF Ab. Transwell experiments revealed that MSC to FLS contact was required for the increase in PlGF production by coculture. Cadherin-11 was expressed both in FLS and MSC, and small interfering RNA knockdown of cadherin-11 in FLS significantly abrogated the enhanced PlGF production under coculture conditions. These data indicate that increased levels of PlGF in RA joints could induce the migration of MSC to the synovium, and interaction of migrated MSC with FLS via cadherin-11 may contribute to angiogenesis and chronic synovitis by enhancing the secretion of PlGF.


Subject(s)
Cadherins/immunology , Cell Communication/immunology , Fibroblasts/immunology , Mesenchymal Stem Cells/immunology , Neovascularization, Pathologic/immunology , Pregnancy Proteins/immunology , Animals , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Blotting, Western , Cadherins/genetics , Cadherins/metabolism , Cells, Cultured , Chemotaxis/drug effects , Chemotaxis/immunology , Coculture Techniques , Collagen , Dose-Response Relationship, Drug , Drug Combinations , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Laminin , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Placenta Growth Factor , Pregnancy Proteins/metabolism , Pregnancy Proteins/pharmacology , Proteoglycans , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Synovial Membrane/immunology , Synovial Membrane/metabolism , Synovial Membrane/pathology , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/immunology , Vascular Endothelial Growth Factor Receptor-1/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL