Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
Heliyon ; 10(6): e27350, 2024 Mar 30.
Article En | MEDLINE | ID: mdl-38496903

Peripheral nerve remodeling and sensitization are involved in cancer-related bone pain. As a member of the transforming growth factor-ß class, bone morphogenetic protein 2 (BMP2) is recognized to have a role in the development of the neurological and skeletal systems. Our previous work showed that BMP2 is critical for bone cancer pain (BCP) sensitization. However, the mechanism remains unknown. In the current study, we demonstrated a substantial increase in BMP2 expression in the dorsal root ganglia (DRG) in a rat model of BCP. Knockdown of BMP2 expression ameliorated BCP in rats. Furthermore, the DRG neurons of rats with BCP expressed higher levels of calcitonin gene-related peptide (CGRP), and BCP was successfully suppressed by intrathecal injection of a CGRP receptor blocker (CGRP8-37). Downregulation of BMP2 expression reduced the expression of CGRP in the DRG of rats with BCP and relieved pain behavior. Moreover, we revealed that upregulation of CGRP expression in the DRG may be induced by activation of the BMPR/Smad1 signaling pathway. These findings suggest that BMP2 contributes to BCP by upregulating CGRP in DRG neurons via activating BMPR/Smad1 signaling pathway and that therapeutic targeting of the BMP2-Smad1-CGRP pathway may ameliorate BCP in the context of advanced cancer.

2.
J Integr Neurosci ; 23(3): 47, 2024 Feb 27.
Article En | MEDLINE | ID: mdl-38538215

BACKGROUND: Bone cancer pain (BCP) is a common primary or metastatic bone cancer complication. Netrin-1 plays an essential role in neurite elongation and pain sensitization. This study aimed to determine the role of netrin-1 from the metastatic bone microenvironment in BCP development and identify the associated signaling pathway for the strategy of BCP management. METHODS: The rat BCP model was established by intratibial implantation of Walker 256 cells. Von Frey filaments measured the mechanical pain threshold. Movement-induced pain was assessed using limb use scores. Expressions of associated molecules in the affected tibias or dorsal root ganglia (DRG) were measured by immunofluorescence, immunohistochemistry, real-time quantitative polymerase chain reaction, or western blotting. Transduction of deleted in colorectal cancer (DCC) signaling was inhibited by intrathecal injection of DCC-siRNA. RESULTS: In BCP rats, the presence of calcitonin gene-related peptide (CGRP)-positive nerve fibers increased in the metastatic bone lesions. The metastatic site showed enrichment of well-differentiated osteoclasts and expressions of netrin-1 and its attractive receptor DCC. Upregulation of DCC and increased phosphorylation levels of focal adhesion kinase (FAK) and Rac family small GTPase 1/Cell division cycle 42 (Rac1/Cdc42) were found in the DRG. Intrathecal administration of DCC-siRNA led to a significant reduction in FAK and Rac1/Cdc42 phosphorylation levels in the DRG, decreased nociceptive nerve innervation, and improved pain behaviors. CONCLUSIONS: Netrin-1 may contribute to the activation of the BCP by inducing nociceptive nerve innervation and improving pain behaviors.


Bone Neoplasms , Cancer Pain , Netrin-1 , Animals , Rats , Bone Neoplasms/complications , Cancer Pain/etiology , DCC Receptor/metabolism , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Netrin-1/genetics , Nociceptors/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , RNA, Small Interfering , Signal Transduction , Tumor Microenvironment , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
3.
J Med Food ; 27(2): 123-133, 2024 Feb.
Article En | MEDLINE | ID: mdl-38100058

Echinacoside (ECH) is a prominent naturally occurring bioactive compound with effects of alleviating myocardial damage. We aimed to explore the beneficial effects of ECH against sepsis-induced myocardial damage and elucidate the potential mechanism. Echocardiography and Masson staining demonstrated that ECH alleviates cardiac function and fibrosis in the cecal ligation and puncture (CLP) model. Transcriptome profiling and network pharmacology analysis showed that there are 51 overlapping targets between sepsis-induced myocardial damage and ECH. Subsequently, chemical carcinogenesis-reactive oxygen species (ROS) were enriched in multiple targets. Wherein, SOD2 may be the potential target of ECH on sepsis-induced myocardial damage. Polymerase chain reaction results showed that ECH administration could markedly increase the expression of SOD2 and reduce the release of ROS. Combined with injecting the inhibitor of SOD2, the beneficial effect of ECH on mortality, cardiac function, and fibrosis was eliminated, and release of ROS was increased after inhibiting SOD2. ECH significantly alleviated myocardial damage in septic mice, and the therapeutic mechanism of ECH is achieved by upregulating SOD2 which decreased the release of ROS.


Glycosides , Myocardium , Sepsis , Mice , Animals , Reactive Oxygen Species , Sepsis/complications , Sepsis/drug therapy , Fibrosis
4.
Front Oncol ; 13: 1031128, 2023.
Article En | MEDLINE | ID: mdl-36910669

Esophageal cancer-related gene-4 (ECRG4), a 148-amino acid propertied and new tumor suppressor, is initially cloned from the normal esophageal epithelium. ECRG4 was found to be expressed not only in esophageal tissues but also in cardiomyocytes. Previous studies demonstrated that ECRG4 is constitutively expressed in esophageal epithelial cells, and its degree of downregulation is directly proportional to prognosis in patients with esophageal cancer. In the heart, ECRG4 shows greater expression in the atria than in the ventricles, which accounts for its heterogeneity. Downregulation of ECRG4 expression level correlates with esophageal cancer, as well as myocardial injuries and arrhythmias. As a result, this review summarizes the possible susceptibility gene, ECRG4 and its associated molecular mechanisms in cancer patients with atrial fibrillation and myocardial injury. The review begins by describing ECRG4's biological background, discusses its expression in the cardiovascular system, lists the clinical and animal research related to the downregulation of ECRG4 in atrial fibrillation, and focuses on its potential role in atrial fibrillation. Downregulation of ECRG4 may increase the risk of atrial fibrillation by affecting ion channels, MMPs expression and inflammatory response. We will then discuss how ECRG4 can be used in the treatment of tumors and arrhythmias, and provide a novel possible strategy to reduce the occurrence of perioperative cardiovascular adverse events in patients with tumors such as esophageal cancer and gastric cancer.

5.
Inflamm Res ; 72(1): 117-132, 2023 Jan.
Article En | MEDLINE | ID: mdl-36346430

BACKGROUND: Neuroinflammation in the peripheral nervous system has been linked to cancer metastasis-induced bone pain. The stimulator of interferon genes (STING), an innate immune sensor for cytosolic DNA, plays an important role in inflammation and cancer metastasis and is reported to be a critical regulator of nociception. Here, we examined the role of STING in primary nociceptive neurons and chronic pain to determine if it could be a new target for treating bone cancer pain (BCP). METHODS: Walker 256 cancer cells were injected intratibially to induce bone cancer pain in rats. STING and its downstream inflammatory factors in dorsal root ganglia (DRG) were detected using western blotting and immunofluorescent staining. Transmission electron microscopy and the BCL2-associated X (Bax) expression were used to detect the mitochondrial stress in DRG neurons. C-176, a specific inhibitor of STING, was used to block STING activation and to test the pain behavior. RESULTS: Mechanical hyperalgesia and spontaneous pain were observed in BCP rats, accompanied by the upregulation of the STING expression in the ipsilateral L4-5 DRG neurons which showed significant mitochondrion stress. The STING/TANK-binding kinase 1 (TBK1)/nuclear factor-kappa B (NF-κB) pathway activation was observed in the DRGs of BCP rats as well as increased IL-1ß, IL-6, and TNF-α expression. C-176 alleviated bone cancer pain and reduced the STING and its downstream inflammatory pathway. CONCLUSION: We provide evidence that STING pathway activation leads to neuroinflammation and peripheral sensitization. Pharmacological blockade of STING may be a promising novel strategy for preventing BCP.


Bone Neoplasms , Cancer Pain , Rats , Animals , Cancer Pain/etiology , Cancer Pain/metabolism , Neuroinflammatory Diseases , Rats, Sprague-Dawley , Pain/etiology , Pain/metabolism , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Hyperalgesia/etiology , Hyperalgesia/metabolism , Ganglia, Spinal
6.
Front Mol Neurosci ; 15: 1026593, 2022.
Article En | MEDLINE | ID: mdl-36385763

Bone cancer pain (BCP) is one of the most common types of pain in cancer patients which compromises the patient's functional status, quality of life, and survival. Central hyperalgesia has increasingly been identified as a crucial factor of BCP, especially in the medial prefrontal cortex (mPFC) which is the main cortical area involved in the process of pain and consequent negative emotion. To explore the genetic changes in the mPFC during BCP occurrence and find possible targets for prediction, we performed transcriptome sequencing of mPFC in the BCP rat model and found a total of 147 differentially expressed mRNAs (DEmRNAs). A protein-protein interaction (PPI) network revealed that the DEmRNAs mainly participate in the inflammatory response. Meanwhile, microglia and astrocytes were activated in the mPFC of BCP rats, further confirming the presence of neuroinflammation. In addition, Gene Ontology (GO) analysis showed that DEmRNAs in the mPFC are mainly involved in antigen processing, presentation of peptide antigen, and immune response, occurring in the MHC protein complex. Besides, the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed that DEmRNAs are mainly enriched in the pathways of phagosome, staphylococcus aureus infection, and antigen processing, in which MHCII participate. Furthermore, immunostaining showed that MHCII is mainly located in the microglia. Microglia are believed to be involved in antigen processing, a key cause of BCP. In vivo, minocycline (MC) treatment inhibits the activation of microglia and reduces the expression of MHCII and proinflammatory cytokines, thereby alleviating BCP and pain-related anxiety. Taken together, our study identified differentially expressed genes in the BCP process and demonstrated that the activation of microglia participates in the inflammatory response and antigen process, which may contribute to BCP.

7.
Cancers (Basel) ; 14(21)2022 Oct 22.
Article En | MEDLINE | ID: mdl-36358605

The medial prefrontal cortex (mPFC) is the main cortical area for processing both sensory and affective aspects of pain. Recently, mPFC was reported to participate in cancer-induced bone pain (CIBP) via the mechanism of central inflammation. STING is a key component of neuroinflammation in the central neuron system by activating downstream TBK1 and NF-κB signaling pathways. We aimed to investigate whether STING regulated neuroinflammation in the mPFC in rat models of CIBP. It is worth noting that we found a significant upregulation of STING in the mPFC after CIBP, accompanied by activation of TBK1 and NF-κB signaling pathways. In addition, pain and anxiety-like behaviors were alleviated by intraperitoneal injection of the STING inhibitor C-176. Furthermore, in microglia GMI-R1 cells, C-176 reversed LPS-induced M1 polarization. Collectively, this evidence indicated that STING may contribute to cancer-induced bone pain by activating TBK1 and NF-κB, and by promoting M1 polarization of microglia in the mPFC.

8.
Front Mol Neurosci ; 15: 963206, 2022.
Article En | MEDLINE | ID: mdl-35979145

Chronic pain interferes with daily functioning and is frequently accompanied by depression. Currently, traditional clinic treatments do not produce satisfactory analgesic effects and frequently result in various adverse effects. Pathogen recognition receptors (PRRs) serve as innate cellular sensors of danger signals, sense invading microorganisms, and initiate innate and adaptive immune responses. Among them, cGAS-STING alerts on the presence of both exogenous and endogenous DNA in the cytoplasm, and this pathway has been closely linked to multiple diseases, including auto-inflammation, virus infection, and cancer. An increasing numbers of evidence suggest that cGAS-STING pathway involves in the chronic pain process; however, its role remains controversial. In this narrative review, we summarize the recent findings on the involvement of the cGAS-STING pathway in chronic pain, as well as several possible mechanisms underlying its activation. As a new area of research, this review is unique in considering the cGAS-STING pathway in sensory neurons and glial cells as a part of a broader understanding of pain, including potential mechanisms of inflammation, immunity, apoptosis, and autophagy. It will provide new insight into the treatment of pain in the future.

9.
Reproduction ; 162(6): 449-460, 2021 10 30.
Article En | MEDLINE | ID: mdl-34718231

The nucleotide binding and oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome plays a critical role in various inflammatory diseases. We sought to investigate the role of NLRP3 inflammasome in uterine activation for labor at term and preterm. We found that NLRP3 inflammasome was activated in the myometrium tissues obtained from the pregnant women undergoing labor at term (TL) compared with those not undergoing labor (TNL) at term. NLRP3 inflammasome was also activated in amnion and chorion-deciduas in TL and preterm labor (PTL) groups. In the mouse model, uterine NLRP3 inflammasome and nuclear factor kappaB (NF-κB) were activated toward term and during labor. Treatment of pregnant mice with lipopolysaccharide (LPS) and RU38486 induced preterm birth (PTB) and also promoted uterine NLRP3 inflammasome and NF-κB activation. Treatment of pregnant mice with NLRP3 inflammasome inhibitor BAY11-7082 and MCC950 delayed the onset of labor and suppressed NLRP3 inflammasome and NF-κB activation in uterus. MCC950 postponed labor onset of the mice with LPS and RU38486 treatment and inhibited NLRP3 inflammasome activation in uterus. Our data provide the evidence that NLRP3 inflammasome is involved in uterine activation for labor onset in term and PTB in humans and mouse model.


Inflammasomes , Premature Birth , Animals , Female , Humans , Infant, Newborn , Mice , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Pregnancy , Uterus/metabolism
10.
Reproduction ; 162(1): 11-20, 2021 05 27.
Article En | MEDLINE | ID: mdl-33929340

Urocortins (UCNs), belonging to corticotropin-releasing hormone (CRH) family, exert their function via CRH receptor type 1 (CRHR1) and 2 (CRHR2). Our previous studies have demonstrated that CRH acts on CRHR1 to potentiate prostaglandins (PGs) output induced by inflammatory stimuli in myometrial cells. In the present study, we sought to investigate the effects of UCNs on prostaglandin (PG) output via CRHR2 in cultured human uterine smooth muscle cells (HUSMCs) from pregnant women at term. We found that UCN and UCN 3 treatment promoted PGE2 and PGF2α secretion in a dose-dependent manner. In contrast, UCN2 dose-dependently inhibited PGE2 and PGF2α secretion. Their effects were reversed by CRHR2 antagonist and CRHR2 siRNA. Mechanically, we showed that UCN and UCN3 suppressed cAMP production and led to Gi activation while UCN2 stimulated cAMP production and activated Gs signaling. Further, UCN and UCN3 but not UCN2 activated NF-κB and MAPK signaling pathways through Gi signaling. UCN and UCN3 stimulation of PGs secretion were dependent on Gi/adenylyl cyclase (AC)/cAMP, NF-κB and MAPK signaling pathways. UCN2 suppression of PGs output was through Gs/AC/cAMP signaling pathways. Our data suggest that UCN, UCN2 and UCN3 can finely regulate PGs secretion via CRHR2, which facilitates the functional status of the uterus during pregnancy.


Dinoprost/metabolism , Dinoprostone/metabolism , Gene Expression Regulation/drug effects , Myometrium/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Urocortins/pharmacology , Uterus/metabolism , Dinoprost/genetics , Female , Humans , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myometrium/drug effects , Receptors, Corticotropin-Releasing Hormone/genetics , Uterus/drug effects
11.
Sheng Li Xue Bao ; 71(2): 248-260, 2019 Apr 25.
Article En | MEDLINE | ID: mdl-31008484

Prostaglandin (PG) E2 plays critical roles during pregnancy and parturition. Emerging evidence indicates that human labour is an inflammatory event. We sought to investigate the effect of PGE2 on the output of proinflammatory cytokines in cultured human uterine smooth muscle cells (HUSMCs) from term pregnant women and elucidate the role of subtypes of PGE2 receptors (EP1, EP2, EP3 and EP4). After drug treatment and/or transfection of each receptor siRNA, the concentrations of inflammatory secreting factors in HUSMCs culture medium were detected by the corresponding ELISA kits. The results showed that, PGE2 increased interleukin 6 (IL-6) and tumor necrosis factor alpha (TNFα) output, decreased chemokine (c-x-c motif) ligand 8 (CXCL8) output in a dose-dependent manner, but had no effect on IL-1ß and chemokine (c-c motif) ligand 2 (CCL-2) secretion of HUSMCs. EP1/EP3 agonist 17-phenyl-trinor-PGE2 stimulated IL-6 and TNFα whilst suppressing IL-1ß and CXCL8 output. The effects of 17-phenyl-trinor-PGE2 on IL-1ß and CXCL8 secretion were remained whereas its effect on IL-6 and TNFα output did not occur in the cells with EP3 knockdown. The stimulatory effects of 17-phenyl-trinor-PGE2 on IL-6 and TNFα were remained whereas the inhibitory effects of 17-phenyl-trinor-PGE2 on IL-1ß secretion was blocked in the cells with EP1 knockdown. Either of EP2 and EP4 agonists stimulated IL-1ß and TNFα output, which was reversed by EP2 and EP4 siRNA, respectively. The inhibitors of phospholipase C (PLC) and protein kinase C (PKC) blocked EP1/EP3 modulation of TNFα and CXCL8 output. PI3K inhibitor LY294002 and P38 inhibitor SB202190 blocked 17-phenyl-trinor-PGE2-induced IL-1ß and IL-6 output, respectively. The inhibitors of adenylyl cyclase and PKA prevented EP2 and EP4 stimulation of IL-1ß and TNFα output, whereas PLC and PKC inhibitors blocked EP2- and EP4-induced TNFα output but not IL-1ß output. Our data suggest that PGE2 receptors exhibit different effects on the output of various cytokines in myometrium, which can subtly modulate the inflammatory microenvironment in myometrium during pregnancy.


Cytokines/metabolism , Myocytes, Smooth Muscle/cytology , Myometrium/cytology , Receptors, Prostaglandin E/physiology , Cells, Cultured , Chromones/pharmacology , Female , Humans , Imidazoles/pharmacology , Inflammation , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases , Pregnancy , Pyridines/pharmacology
12.
Reproduction ; 153(5): 535-543, 2017 05.
Article En | MEDLINE | ID: mdl-28188160

Recent evidence suggests that uterine activation for labor is associated with inflammation within uterine tissues. Hydrogen sulfide (H2S) plays a critical role in inflammatory responses in various tissues. Our previous study has shown that human myometrium produces H2S via its generating enzymes cystathionine-γ-lyase (CSE) and cystathionine-ß-synthetase (CBS) during pregnancy. We therefore explored whether H2S plays a role in the maintenance of uterine quiescence during pregnancy. Human myometrial biopsies were obtained from pregnant women at term. Uterine smooth muscle cells (UMSCs) isolated from myometrial tissues were treated with various reagents including H2S. The protein expression of CSE, CBS and contraction-associated proteins (CAPs) including connexin 43, oxytocin receptor and prostaglandin F2α receptor determined by Western blot. The levels of cytokines were measured by ELISA. The results showed that CSE and CBS expression inversely correlated to the levels of CAPs and activated NF-κB in pregnant myometrial tissues. H2S inhibited the expression of CAPs, NF-κB activation and the production of interleukin (IL)-1ß, IL-6 and tumor necrosis factor α (TNFα) in cultured USMCs. IL-1ß treatment reversed H2S inhibition of CAPs. Knockdown of CSE and CBS prevented H2S suppression of inflammation. H2S modulation of inflammation is through KATP channels and phosphoinositide 3-kinase (PI3K) and extracellular signal-regulated kinase (ERK) signaling pathways. H2S activation of PI3K and ERK signaling is dependent on KATP channels. Our data suggest that H2S suppresses the expression of CAPs via inhibition of inflammation in myometrium. Endogenous H2S is one of the key factors in maintenance of uterine quiescence during pregnancy.


Air Pollutants/pharmacology , Hydrogen Sulfide/pharmacology , Inflammation/drug therapy , Labor, Obstetric/drug effects , Uterine Contraction/drug effects , Uterus/drug effects , Cells, Cultured , Cystathionine gamma-Lyase/metabolism , Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Inflammation/metabolism , Labor, Obstetric/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myometrium/cytology , Myometrium/drug effects , Myometrium/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pregnancy , Signal Transduction/drug effects , Uterine Contraction/metabolism , Uterus/metabolism
13.
PLoS One ; 11(4): e0152838, 2016.
Article En | MEDLINE | ID: mdl-27035826

A major cause of preterm labor in pregnant women is intra-amniotic infection, which is mediated by an inflammatory process. Hydrogen sulfide (H2S), a gaseous transmitter, has been implicated to be involved in inflammatory responses. We sought to investigate whether H2S affects infectious preterm birth using the mouse model of lipopolysaccharides (LPS)-induced preterm birth. Administration of LPS at 0.4 mg/kg with two injections intraperitoneally (i.p.) on gestational day 14.5 induced preterm labor. LPS significantly increased leukocyte infiltration in uterus, stimulated the expression of pro-inflammatory cytokines interleukin 1ß (IL-1ß), IL-6, tumor necrosis factor α (TNF-α), CCL2 and CXCL15 in myometrium. Administration of NaHS (i.p.) delayed the onset of labor induced by LPS in a dose-dependent manner. NaHS prevented leukocyte infiltration into intrauterine tissues and inhibited the production of pro-inflammatory cytokines in myometrium and decreased the levels of these cytokines in maternal circulation. H2S also decreased LPS-activated extracellular signal-regulated kinase (ERK) 1/2/ nuclear factor (NF)-κB signaling pathways in myometrium. This study provides new in vivo evidence for the roles of H2S in attenuating inflammation, and a potential novel therapeutic strategy for infection-related preterm labor.


Cytokines/metabolism , Hydrogen Sulfide/pharmacology , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Obstetric Labor, Premature/chemically induced , Animals , Female , Inflammation/metabolism , Mice , Mice, Inbred BALB C , Pregnancy
14.
Mol Hum Reprod ; 21(7): 603-14, 2015 Jul.
Article En | MEDLINE | ID: mdl-25882540

Prostaglandin F2α (PGF2α) plays a critical role in the initiation and process of parturition. Since human labor has been described as an inflammatory event, we investigated the role of PGF2α in the inflammatory process using cultured human uterine smooth muscle cells (HUSMCs) isolated from term pregnant women as a model. Using a multiplex assay, HUSMCs treated with PGF2α changed their output of a number of cytokines and chemokines, with a distinct response pattern that differed between HUSMCs isolated from the upper and lower segment region of the uterus. Confirmatory enzyme-linked immunosorbent assays (ELISAs) showed that PGF2α stimulated increased output of interleukin (IL) 1ß, IL6, IL8 (CXCL8) and monocyte chemotactic protein-1 (MCP1, also known as chemokine (c-c motif) ligand 2, CCL2) by HUSMCs isolated from both upper and lower uterine segments. In contrast, PGF2α inhibited tumor necrosis factor α (TNFα) release by HUMSCs from the lower uterine segment while the output of TNFα was undetectable in the upper segment. Small interfering (si) RNA mediated knockdown of the PGF2α receptor prevented the changes in cytokine and chemokine output by the HUSMCs. Since the PGF2α receptor (PTGFR) couples via the Gq protein and subsequently activates the phospholipase C (PLC) and protein kinase C (PKC) signaling pathways, we examined the role of these pathways in PGF2α modulation of the cytokines. Inhibition of PLC and PKC reversed the effects of PGF2α. PGF2α activated multiple signaling pathways including extracellular signal-regulated kinases (ERK) 1/2, phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K), P38, calcineurin/nuclear factor of activated T-cells (NFAT) and NF-κB signaling. Inhibition of ERK reversed PGF2α-induced IL1ß, IL6 and CCL2 output, while inhibition of PI3K blocked the effect of PGF2α on IL6, CXCL8 and CCL2 output and inhibition of NF-κB reversed PGF2α-induced IL1ß and CCL2 output. NFAT was involved in PGF2α modulation of CCL2 and TNFα output. In conclusion, our results support a role of PGF2α in creating an inflammatory environment during the late stage of human pregnancy.


Chemokines/metabolism , Cytokines/metabolism , Dinoprost/pharmacology , Myocytes, Smooth Muscle/drug effects , Myometrium/drug effects , Signal Transduction/drug effects , Female , Humans , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Myometrium/cytology , Myometrium/metabolism , Pregnancy , Signal Transduction/physiology
15.
Reproduction ; 149(1): 139-46, 2015 Jan.
Article En | MEDLINE | ID: mdl-25342173

Prostaglandin F2α (PGF2A) has multiple roles in the birth process in addition to its vital contractile role. Our previous study has demonstrated that PGF2A can modulate uterine activation proteins (UAPs) in cultured pregnant human myometrial smooth muscle cells (HMSMCs). The objective of this study was to define the signalling pathways responsible for PGF2A modulation of UAPs in myometrium. It was found that PGF2A stimulated the expression of (GJA1) connexin 43 (CX43), prostaglandin endoperoxide synthase 2 (PTGS2) and oxytocin receptor (OTR) in cultured HMSMCs. The inhibitors of phospholipase C (PLC) and protein kinase C (PKC) blocked PGF2A-stimulated expression of CX43. The inhibitors of ERK, P38 and NFκB also blocked the effect of PGF2A on CX43 expression, whereas PI3K and calcineurin/nuclear factor of activated T-cells (NFAT) pathway inhibitors did not reverse the effect of PGF2A on CX43. For PTGS2 and OTR, PLC, PI3K, P38 and calcineurin/NFAT signalling pathways were involved in PGF2A action, whereas PKC and NFκB signalling were not involved. In addition, PGF2A activated NFAT, PI3K, NFκB, ERK and P38 signalling pathways. Our data suggest that PGF2A stimulates CX43, PTGS2 and OTR through divergent signalling pathways.


Connexin 43/metabolism , Cyclooxygenase 2/metabolism , Dinoprost/pharmacology , Gene Expression Regulation/drug effects , Receptors, Oxytocin/metabolism , Signal Transduction/drug effects , Uterus/metabolism , Cells, Cultured , Female , Humans , Immunoblotting , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myometrium/cytology , Myometrium/drug effects , Myometrium/metabolism , Oxytocics/pharmacology , Phosphorylation/drug effects , Pregnancy , Uterus/cytology , Uterus/drug effects
16.
Mol Med Rep ; 11(4): 2513-9, 2015 Apr.
Article En | MEDLINE | ID: mdl-25435100

Autophagy is a highly conserved pathway that permits recycling of nutrients within the cell and is rapidly upregulated during starvation or cell stress. Autophagy has been implicated in the pathophysiological process of warm ischemia­reperfusion injury in the rat lung. Cold ischemia (CI) preservation for lung transplantation also exhibits cell stress and nutrient deprivation, however, little is known with regard to the involvement of autophagy in this process. In the present study, CI preservation­induced autophagy and apoptosis was investigated in the lungs of Sprague Dawley rats. Sprague Dawley rat lungs were flushed and preserved at 4˚C (i.e. CI) for various durations (0, 3, 6, 12 and 24 h). The levels of autophagy, autophagic cell death and apoptosis were measured at each time point following CI. The results revealed that autophagy was induced by CI preservation, which was initiated at 3 h, peaked at 6 h after CI and declined thereafter. Additionally, a coexistence of autophagic cell death and apoptosis was observed in rat lung tissues following prolonged CI. These findings demonstrate that autophagy is involved in the pathophysiological process of lung CI. Furthermore, autophagic cell death in addition to necrosis and apoptosis occurs following CI in the lung. CI preservation may therefore be a potential mechanism of lung injury during organ preservation prior to lung transplantation.


Autophagy , Cold Ischemia , Lung/metabolism , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy/genetics , Beclin-1 , Lung Transplantation , Male , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Models, Animal , Organ Preservation , Rats
17.
J Clin Endocrinol Metab ; 99(2): E199-208, 2014 Feb.
Article En | MEDLINE | ID: mdl-24248185

CONTEXT: An increasing body of evidence indicates that human labor, either term or preterm, is an inflammatory event. CRH has been implicated to be a trigger of human parturition. OBJECTIVE: To investigate whether CRH induces the cascades of inflammation in human pregnant myometrium, thereby leading to activation of uterus. DESIGN: The myometrial tissues were obtained from pregnant women who were in labor or not in labor at term. The output of cytokines and prostaglandins (PGs) was determined by Multiplex and ELISA. Western blot analysis was used to determine the levels of uterine activation proteins (UAPs). RESULTS: The levels of chemokines and cytokines as well as activated nuclear factor-κB (NF-κB) were increased in the term labor group more than the not term labor group. CRH stimulated production of a number of chemokines and cytokines in cultured uterine smooth muscle cells (USMCs), which induced chemotaxis of monocytes. These effects were mediated by CRH receptor 1 (CRHR1) and dependent on adenylyl cyclase/protein kinase (PKA) and NF-κB signaling. Cocultures of CRH-treated USMCs with monocytes greatly enhanced the output of cytokines and chemokines as well as PGs in cultures and increased the expression of uterine activation proteins (UAPs) in USMCs. IL-1ß, IL-6, and TNF-α stimulated the expression of UAPs and output of PGs in USMCs. CONCLUSIONS: CRH induces the production of chemokines and cytokines in myometrium at term and subsequently results in the cascade of inflammation in uterus. The inflammation induced by CRH can lead to activation of uterus.


Chemokines/metabolism , Corticotropin-Releasing Hormone/pharmacology , Cytokines/metabolism , Inflammation/metabolism , Myometrium/drug effects , Parturition/metabolism , Adult , Cells, Cultured , Corticotropin-Releasing Hormone/metabolism , Female , Humans , Labor, Obstetric/metabolism , Myometrium/cytology , Myometrium/metabolism , NF-kappa B/metabolism , Pregnancy
18.
Am J Pathol ; 182(5): 1585-94, 2013 May.
Article En | MEDLINE | ID: mdl-23506845

The chorion laeve controls the levels of active prostaglandins within the uterus by NAD-dependent 15-hydroxy prostaglandin dehydrogenase (PGDH). The expression of PGDH in chorion is modulated by glucocorticoids and progesterone. In this study, we investigated glucocorticoid receptor (GR) and progesterone receptor A and B (PRA and PRB) in the regulation of PGDH expression in chorion, and we determined whether reduced PGDH expression in chorion during labor is associated with the changes in GR and PR expression by real-time RT-PCR and Western blot analysis. Dexamethasone (DEX) inhibited PGDH expression whereas progesterone stimulated PGDH expression in chorionic trophoblasts. DEX suppressed PGDH expression in GR overexpression and PR knockdown cells. The inhibitory effect of DEX did not occur in GR knockdown cells. Progesterone inhibited PGDH in GR overexpression and PR knockdown cells and it stimulated PGDH in PRB overexpression cells whereas it suppressed PGDH in PRA overexpression cells. Knockdown of c-Jun resulted in a loss of progesterone- and DEX-induced effects. PGDH was down-regulated in chorion tissues during labor. PRB was decreased whereas PRA and GR were increased in chorion during labor. Glucocorticoids inhibit PGDH expression via GR in chorionic trophoblasts. Progesterone enhances PGDH expression through PRB, whereas it inhibits PGDH expression via GR and PRA. Decreased PGDH expression is associated with increased GR and PRA, although decreased PRB, in chorion during labor.


Chorion/enzymology , Hydroxyprostaglandin Dehydrogenases/metabolism , Labor, Obstetric/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Progesterone/metabolism , Cells, Cultured , Chorion/cytology , Chorion/drug effects , Dexamethasone/pharmacology , Dihydrotestosterone/analogs & derivatives , Dihydrotestosterone/pharmacology , Extraembryonic Membranes/drug effects , Extraembryonic Membranes/metabolism , Female , Fluorescent Antibody Technique , Glucocorticoids/pharmacology , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Labor, Obstetric/drug effects , Pregnancy , Progesterone/pharmacology , Proto-Oncogene Proteins c-jun/metabolism , Transcription, Genetic/drug effects , Trophoblasts/cytology , Trophoblasts/drug effects , Trophoblasts/metabolism , Up-Regulation/drug effects
19.
J Surg Res ; 182(1): e25-33, 2013 Jun 01.
Article En | MEDLINE | ID: mdl-23122581

BACKGROUND: Lung injury induced by ischemia or reperfusion significantly accounts for the risk of early mortality of lung transplantation (LT). Recent studies have demonstrated that hydrogen sulfide (H2S) and its endogenous synthase cystathionine-γ-lyase (CSE) confer protection against injury induced by ischemia or reperfusion in various organs. This prompted us to define the role of CSE/H2S pathway in transplantation-induced lung injury. METHODS: We performed single left LT in male Sprague-Dawley rats after 3 h of cold ischemia time. H2S donor NaHS (14 µmol/kg, intraperitoneally) or CSE inhibitor propargylglycine (37.5 mg/kg, intraperitoneally) was administered 15 min before the start of the LT. CSE protein expression, H2S generation, and the severity of pulmonary graft injuries were estimated at 24 h after reperfusion. RESULTS: Both CSE protein expression and H2S generation were markedly decreased in transplanted rat lungs compared with those in sham-operated lungs. In the lung-transplanted rats, NaHS administration significantly improved pulmonary function and decreased lipid peroxidation and myeloperoxidase activity. In addition, NaHS inhibited the production of interleukin 1ß but increased interleukin 10 levels in graft lung tissues. In contrast, propargylglycine further exacerbated pulmonary function and lung injuries after experimental orthotopic LT. CONCLUSIONS: To our knowledge, this study for the first time has demonstrated that the suppression of CSE expression and H2S production is associated with transplantation-induced lung injury. Both exogenous and endogenous H2S seem to have protective effects against acute LT injury by their multiple functions including antioxidation and anti-inflammation, suggesting that modulation of H2S levels may be considered a potential therapeutic approach in LT.


Cold Ischemia/adverse effects , Hydrogen Sulfide/antagonists & inhibitors , Lung Injury/etiology , Lung Injury/metabolism , Lung Transplantation , Reperfusion Injury/complications , Alkynes/pharmacology , Animals , Cystathionine gamma-Lyase/antagonists & inhibitors , Cystathionine gamma-Lyase/drug effects , Cystathionine gamma-Lyase/metabolism , Enzyme Inhibitors/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , Hydrogen Sulfide/metabolism , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Lung Injury/physiopathology , Male , Models, Animal , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Sulfides/pharmacology
20.
J Clin Endocrinol Metab ; 97(10): E1851-61, 2012 Oct.
Article En | MEDLINE | ID: mdl-22869609

CONTEXT: Our previous study has demonstrated that CRH has differential effects on human uterine contractility before and after onset of labor. Intracellular Ca2+ concentration ([Ca2+]i) mobilization plays an important role in the control of uterine contraction. OBJECTIVE: Our objective was to investigate the effects of CRH on [Ca2+]i homeostasis in laboring and nonlaboring myometrial cells and determine subsequent signaling involved in [Ca2+]i regulation by CRH. DESIGN: The myometrial tissues were obtained from pregnant women who were undergoing or not undergoing labor at term. [Ca2+]i was determined by Ca2+ imaging system using the fluorescent dye fura-2-acetoxymethyl ester. Western blot analysis, ELISA, and RIA were used to determine the signaling pathways induced by CRH. RESULTS: CRH induced Ca2+ transient in laboring cells, which was blocked by CRH receptor type 1 (CRHR1) antagonist antalarmin. CRHR1 knockdown impaired this effect of CRH. CRH activated Gi protein, decreased cAMP production, and induced phosphorylated phospholipase C-ß3 and inositol-1,4,5-triphosphate production. Phospholipase C and inositol-1,4,5-triphosphate receptor inhibitors blocked the CRH-induced Ca2+ transient in laboring cells. CRH did not induce whereas antalarmin induced the Ca2+ transient in nonlaboring cells. Knockdown of CRHR1 impaired the effect of antalarmin. CRH acted on CRHR1 to activate Gs in nonlaboring cells. Forskolin blocked antalarmin-induced Ca2+ transient. CONCLUSIONS: CRH acts on CRHR1 to activate different signaling pathways before and after onset of labor, thereby resulting in differential calcium signaling in response to CRH. The signaling pathways of CRHR1 might serve as a target for the development of new therapeutic strategies for preterm birth.


Calcium Signaling/physiology , Corticotropin-Releasing Hormone/metabolism , Labor, Obstetric/metabolism , Myometrium/metabolism , Uterine Contraction/physiology , Calcium/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Female , Fluorescent Dyes , Fura-2/analogs & derivatives , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Homeostasis/physiology , Humans , Myometrium/cytology , Pregnancy , Premature Birth/metabolism , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/metabolism
...