Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-38961846

RESUMO

The prostaglandin E2 (PGE2) receptor EP3 has been detected in the thick ascending limb (TAL) and the collecting duct of the kidney, where its actions are proposed to inhibit water reabsorption. However, EP3 is also expressed in other cell types, including vascular endothelial cells. The aim here was to determine the contribution of EP3 in renal water handling in male and female adult mice by phenotyping a novel mouse model with doxycycline-dependent deletion of EP3 throughout the kidney tubule (EP3-/- mice). RNAscope demonstrated that EP3 was highly expressed in the cortical and medullary TAL of adult mice. Compared to controls EP3 mRNA expression was reduced by >80% in whole kidney (RT-qPCR) and non-detectable (RNAscope) in renal tubules of EP3-/- mice. Under basal conditions, there were no significant differences in control and EP3-/- mice of both genders in food and water intake, bodyweight, urinary output or clinical biochemistries. No differences were detectable between genotypes in handling of an acute water load, or in their response to the vasopressin analogue dDAVP. No differences in water handling were observed when PGE2 production was enhanced using a 1% NaCl load. Expression of proteins involved in kidney water handling were not different between genotypes. This study demonstrates that renal tubular EP3 is not essential for body fluid homeostasis in males or females, even when PGE2 levels are high. The mouse model is a novel tool for examining the role of EP3 in kidney function independently of potential developmental abnormalities or systemic effects.

2.
J Leukoc Biol ; 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-39041661

RESUMO

Tumor-derived prostaglandin E2 (PGE2) impairs anti-tumor immunity by priming suppressive functions on various immune cell types, including dendritic cells (DCs). In this way, tumors mediate DC dysfunction and hamper their anti-tumoral activity. PGE2 is known to modulate DC function via signaling through the E-prostanoid receptor type (EP) 2 and EP4. Preclinical studies have demonstrated the therapeutic value of targeting EP2/4 receptor signaling in DCs. Ongoing phase I clinical trials with EP antagonists have shown immunomodulation in cancer patients. However, the systemic drug administration leads to off-target events and subsequent side-effects. To limit the off-target effects of EP targeting, EP2 and EP4 antagonists were encapsulated in polymeric nanoparticles (NPs). In this study we evaluated the efficacy of EP2/4 specific antagonists encapsulated in NPs to protect cDC2s from suppressive effects of tumor-derived PGE2 in different tumor models. We show that tumor-derived PGE2 signals via EP2/4 to mediate the acquisition of a suppressive phenotype of cDC2s. EP2/4 antagonists encapsulated NPs impaired the conversion of cDC2s towards a suppressive state and inhibited the occurrence of suppressive features such as IL-10 production or the ability to expand Tregs. Importantly, the NPs abolished the transition towards this suppressive state in different tumor models: Melanoma-conditioned media, ascites fluid derived from ovarian cancer patients (2D), and upon coculture with colorectal cancer patient-derived organoids (3D). We propose that targeting the PGE2-EP2/4 axis using NPs can achieve immunomodulation in the immune system of cancer patients, alleviate tumor-derived suppression, and thus facilitate the development of potent anti-tumor immunity in cancer patients.

3.
Cell Rep ; 43(3): 113893, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38446662

RESUMO

Prostaglandin F2α (PGF2α) and thromboxane A2 (TXA2) are endogenous arachidonic acid metabolites, modulating diverse physiological processes including inflammation and cardiovascular homeostasis through activating PGF2α receptor (FP) and TXA2 receptor (TP). Ligands targeting FP and TP have demonstrated efficacy in treating conditions like glaucoma and cardiovascular diseases in humans, as well as reproductive-related diseases in animals. Here, we present five cryoelectron microscopy structures illustrating FP and TP in complex with Gq and bound to PGF2α (endogenous ligand), latanoprost acid (a clinical drug), and two other synthetic agonists. Combined with mutational and functional studies, these structures reveal not only structural features for the specific recognition of endogenous ligands and attainment of receptor selectivity of FP and TP but also the common mechanisms of receptor activation and Gq protein coupling. The findings may enrich our knowledge of ligand recognition and signal transduction of the prostanoid receptor family and facilitate rational ligand design toward these two receptors.


Assuntos
Receptores de Prostaglandina , Transdução de Sinais , Humanos , Animais , Ligantes , Microscopia Crioeletrônica , Receptores de Prostaglandina/metabolismo , Prostaglandinas
4.
Front Immunol ; 15: 1355769, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38343540

RESUMO

Tumors educate their environment to prime the occurrence of suppressive cell subsets, which enable tumor evasion and favors tumor progression. Among these, there are the myeloid-derived suppressor cells (MDSCs), their presence being associated with the poor clinical outcome of cancer patients. Tumor-derived prostaglandin E2 (PGE2) is known to mediate MDSC differentiation and the acquisition of pro-tumor features. In myeloid cells, PGE2 signaling is mediated via E-prostanoid receptor type 2 (EP2) and EP4. Although the suppressive role of PGE2 is well established in MDSCs, the role of EP2/4 on human MDSCs or whether EP2/4 modulation can prevent MDSCs suppressive features upon exposure to tumor-derived PGE2 is poorly defined. In this study, using an in vitro model of human monocytic-MDSCs (M-MDSCs) we demonstrate that EP2 and EP4 signaling contribute to the induction of a pro-tumor phenotype and function on M-MDSCs. PGE2 signaling via EP2 and EP4 boosted M-MDSC ability to suppress T and NK cell responses. Combined EP2/4 blockade on M-MDSCs during PGE2 exposure prevented the occurrence of these suppressive features. Additionally, EP2/4 blockade attenuated the suppressive phenotype of M-MDSCs in a 3D coculture with colorectal cancer patient-derived organoids. Together, these results identify the role of tumor-derived PGE2 signaling via EP2 and EP4 in this human M-MDSC model, supporting the therapeutic value of targeting PGE2-EP2/4 axis in M-MDSCs to alleviate immunosuppression and facilitate the development of anti-tumor immunity.


Assuntos
Células Supressoras Mieloides , Humanos , Células Supressoras Mieloides/metabolismo , Dinoprostona/metabolismo , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Monócitos
5.
Eur J Immunol ; 54(3): e2350770, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38088451

RESUMO

Dendritic cells (DCs) shape adaptive immunity in response to environmental cues such as cytokines or lipid mediators, including prostaglandin E2 (PGE2). In cancer, tumors are known to establish an enriched PGE2 microenvironment. Tumor-derived PGE2 primes regulatory features across immune cells, including DCs, facilitating tumor progression. PGE2 shapes DC function by providing signaling via its two so-called E-prostanoid receptors (EPs) EP2 and EP4. Although studies with monocyte-derived DCs have shown the importance of PGE2 signaling, the role of PGE2-EP2/EP4 on conventional DCs type 2 (cDC2s), is still poorly defined. In this study, we investigated the function of EP2 and EP4 using specific EP antagonists on human cDC2s. Our results show that EP2 and EP4 exhibit different functions in cDC2s, with EP4 modulating the upregulation of activation markers (CD80, CD86, CD83, MHC class II) and the production of IL-10 and IL-23. Furthermore, PGE2-EP4 boosts CCR type 7-based migration as well as a higher T-cell expansion capacity, characterized by the enrichment of suppressive rather than pro-inflammatory T-cell populations. Our findings are relevant to further understanding the role of EP receptors in cDC2s, underscoring the benefit of targeting the PGE2-EP2/4 axis for therapeutic purposes in diseases such as cancer.


Assuntos
Dinoprostona , Neoplasias , Humanos , Linfócitos T , Receptores de Prostaglandina E Subtipo EP2 , Receptores de Prostaglandina E Subtipo EP4 , Microambiente Tumoral
6.
Front Immunol ; 14: 1276852, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37942331

RESUMO

Introduction: The intestinal barrier plays a crucial role in distinguishing foods from toxins. Prostaglandin D2 (PGD2) is one of the lipid-derived autacoids synthesized from cell membrane-derived arachidonic acid. We previously reported that pharmacological stimulation of PGD2 receptor, D prostanoid 1 (DP1) attenuated the symptoms of azoxymethane/dextran sodium sulfate-induced colitis and ovalbumin-induced food allergy in mouse models. These observations suggested that DP1 stimulation protects the intestinal barrier. The present study aimed to uncover the effects of DP1 stimulation on intestinal barrier function and elucidate the underlying mechanisms. Materials and methods: Intestinal permeability was assessed in mice by measuring the transfer of orally administered fluorescein isothiocyanate-dextran (40 kDa) into the blood. The DP1 agonist BW245C (1 mg/kg) was administered 10 min prior to dextran administration. The intestinal permeability was confirmed using the ex vivo everted sac method. Tight junction integrity was evaluated in vitro by measuring the transepithelial electrical resistance (TER) in the human intestinal epithelial cell line Caco-2. Mucus secretion was assessed by observing Alcian Blue-stained intestinal sections. Results: Pharmacological DP1 stimulation reduced intestinal permeability both in vivo and ex vivo. Immunohistochemical staining showed that DP1 was strongly expressed on the apical side of the epithelial cells. DP1 stimulation did not affect TER in vitro but induced mucus secretion from goblet cells. Mucus removal by a mucolytic agent N-acetyl-l-cysteine canceled the inhibition of intestinal permeability by DP1 stimulation. Conclusion: These observations suggest that pharmacological DP1 stimulation decreases intestinal permeability by stimulating mucus secretion.


Assuntos
Dextranos , Prostaglandinas , Humanos , Animais , Camundongos , Prostaglandina D2/metabolismo , Células CACO-2 , Muco/metabolismo , Permeabilidade
7.
Eur J Pharmacol ; 956: 175963, 2023 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-37543159

RESUMO

Prostaglandin (PG) D2, a commonly considered vasodilator through D prostanoid receptor-1 (DP1), might also evoke vasoconstriction via acting on the thromboxane (Tx)-prostanoid receptor (the original receptor of TxA2; TP) and/or E prostanoid receptor-3 (one of the vasoconstrictor receptors of PGE2; EP3). This study aimed to test the above hypothesis in the mouse renal vascular bed (main renal arteries and perfused kidneys) and/or mesenteric resistance arteries and determine how the vasoconstrictor mechanism influences the overall PGD2 effect on systemic blood pressure under in vivo conditions. Experiments were performed on control wild-type (WT) mice and mice with deficiencies in TP (TP-/-) and/or EP3 (EP3-/-). Here we show that PGD2 indeed evoked vasoconstrictor responses in the above-mentioned tissues of WT mice, which were however not only reduced by TP-/- or EP3-/-, but also reversed by TP-/-/EP3-/- in some of the above tissues (mesenteric resistance arteries or perfused kidneys) to dilator reactions that were reduced by non-selective DP antagonism. A slight or mild pressor response was also observed with PGD2 under in vivo conditions, and this was again reversed to a depressor response in TP-/- or TP-/-/EP3-/- mice. Non-selective DP antagonism reduced the PGD2-evoked depressor response in TP-/-/EP3-/- mice as well. These results thus demonstrate that like other PGs, PGD2 activates TP and/or EP3 to evoke vasoconstrictor activities, which can outweigh its concurrent vasodepressor activity mediated mainly through DP1, and hence result in a pressor response, although the response might only be of a slight or mild extent.


Assuntos
Prostaglandinas , Vasoconstritores , Camundongos , Animais , Tromboxanos , Receptores de Tromboxanos , Receptores de Prostaglandina E Subtipo EP3 , Receptores de Prostaglandina , Prostaglandina D2/farmacologia
8.
Transl Neurodegener ; 12(1): 32, 2023 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-37337289

RESUMO

Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by selective degeneration of upper and lower motor neurons. The pathogenesis of ALS remains largely unknown; however, inflammation of the spinal cord is a focus of ALS research and an important pathogenic process in ALS. Prostaglandin E2 (PGE2) is a major lipid mediator generated by the arachidonic-acid cascade and is abundant at inflammatory sites. PGE2 levels are increased in the postmortem spinal cords of ALS patients and in ALS model mice. Beneficial therapeutic effects have been obtained in ALS model mice using cyclooxygenase-2 inhibitors to inhibit the biosynthesis of PGE2, but the usefulness of this inhibitor has not yet been proven in clinical trials. In this review, we present current evidence on the involvement of PGE2 in the progression of ALS and discuss the potential of microsomal prostaglandin E synthase (mPGES) and the prostaglandin receptor E-prostanoid (EP) 2 as therapeutic targets for ALS. Signaling pathways involving prostaglandin receptors mediate toxic effects in the central nervous system. In some situations, however, the receptors mediate neuroprotective effects. Our recent studies demonstrated that levels of mPGES-1, which catalyzes the final step of PGE2 biosynthesis, are increased at the early-symptomatic stage in the spinal cords of transgenic ALS model mice carrying the G93A variant of superoxide dismutase-1. In addition, in an experimental motor-neuron model used in studies of ALS, PGE2 induces the production of reactive oxygen species and subsequent caspase-3-dependent cytotoxicity through activation of the EP2 receptor. Moreover, this PGE2-induced EP2 up-regulation in motor neurons plays a role in the death of motor neurons in ALS model mice. Further understanding of the pathophysiological role of PGE2 in neurodegeneration may provide new insights to guide the development of novel therapies for ALS.


Assuntos
Esclerose Lateral Amiotrófica , Doenças Neurodegenerativas , Camundongos , Animais , Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Doenças Neurodegenerativas/metabolismo , Neurônios Motores/patologia , Camundongos Transgênicos , Dinoprostona/metabolismo , Dinoprostona/farmacologia , Dinoprostona/uso terapêutico
9.
Int J Mol Sci ; 24(9)2023 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-37176081

RESUMO

Sphingolipids are important biological mediators both in health and disease. We investigated the vascular effects of enhanced sphingomyelinase (SMase) activity in a mouse model of type 2 diabetes mellitus (T2DM) to gain an understanding of the signaling pathways involved. Myography was used to measure changes in the tone of the thoracic aorta after administration of 0.2 U/mL neutral SMase in the presence or absence of the thromboxane prostanoid (TP) receptor antagonist SQ 29,548 and the nitric oxide synthase (NOS) inhibitor L-NAME. In precontracted aortic segments of non-diabetic mice, SMase induced transient contraction and subsequent weak relaxation, whereas vessels of diabetic (Leprdb/Leprdb, referred to as db/db) mice showed marked relaxation. In the presence of the TP receptor antagonist, SMase induced enhanced relaxation in both groups, which was 3-fold stronger in the vessels of db/db mice as compared to controls and could not be abolished by ceramidase or sphingosine-kinase inhibitors. Co-administration of the NOS inhibitor L-NAME abolished vasorelaxation in both groups. Our results indicate dual vasoactive effects of SMase: TP-mediated vasoconstriction and NO-mediated vasorelaxation. Surprisingly, in spite of the general endothelial dysfunction in T2DM, the endothelial NOS-mediated vasorelaxant effect of SMase was markedly enhanced.


Assuntos
Diabetes Mellitus Tipo 2 , Óxido Nítrico Sintase Tipo III , Camundongos , Animais , Óxido Nítrico Sintase Tipo III/metabolismo , Vasodilatação , Esfingomielina Fosfodiesterase/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , NG-Nitroarginina Metil Éster/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Óxido Nítrico/metabolismo , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/metabolismo
10.
FASEB J ; 36(11): e22576, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36183332

RESUMO

G protein-coupled receptors (GPCRs) are widely therapeutically targeted, and recent advances in allosteric modulator development at these receptors offer further potential for exploitation. Intracellular allosteric modulators (IAM) represent a class of ligands that bind to the receptor-effector interface (e.g., G protein) and inhibit agonist responses noncompetitively. This potentially offers greater selectivity between receptor subtypes compared to classical orthosteric ligands. However, while examples of IAM ligands are well described, a more general methodology for assessing compound interactions at the IAM site is lacking. Here, fluorescent labeled peptides based on the Gα peptide C terminus are developed as novel binding and activation biosensors for the GPCR-IAM site. In TR-FRET binding studies, unlabeled peptides derived from the Gαs subunit were first characterized for their ability to positively modulate agonist affinity at the ß2 -adrenoceptor. On this basis, a tetramethylrhodamine (TMR) labeled tracer was synthesized based on the 19 amino acid Gαs peptide (TMR-Gαs19cha18, where cha = cyclohexylalanine). Using NanoBRET technology to detect binding, TMR-Gαs19cha18 was recruited to Gs coupled ß2 -adrenoceptor and EP2 receptors in an agonist-dependent manner, but not the Gi-coupled CXCR2 receptor. Moreover, NanoBRET competition binding assays using TMR-Gαs19cha18 enabled direct assessment of the affinity of unlabeled ligands for ß2 -adrenoceptor IAM site. Thus, the NanoBRET platform using fluorescent-labeled G protein peptide mimetics offers novel potential for medium-throughput screens to identify IAMs, applicable across GPCRs coupled to a G protein class. Using the same platform, Gs peptide biosensors also represent useful tools to probe orthosteric agonist efficacy and the dynamics of receptor activation.


Assuntos
Técnicas Biossensoriais , Receptores de Interleucina-8B , Regulação Alostérica , Sítio Alostérico , Aminoácidos , Proteínas de Ligação ao GTP/metabolismo , Ligantes , Peptídeos/metabolismo , Peptídeos/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Interleucina-8B/metabolismo
11.
Biol Pharm Bull ; 45(8): 992-997, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35908909

RESUMO

Prostanoids are a group of typical lipid mediators that are biosynthesized from arachidonic acid by the actions of cyclooxygenases and their subsequent terminal synthases. Prostanoids exert a wide variety of actions through their specific membrane receptors on target cells. In addition to their classical actions, including fever, pain, and inflammation, prostanoids have been shown to play pivotal roles in various biological processes, such as female reproduction and the maintenance of vascular and gut homeostasis. Moreover, recent research using mice deficient in each of the prostanoid receptors, or using agonists/antagonists specific for each receptor clarified novel actions of prostanoids that had long been unknown, and the mechanisms therein. In this review, we introduce recent advances in the fields of metabolic control by prostanoid receptors such as in adipocyte differentiation, lipolysis, and adipocyte browning in adipose tissues, and discuss the potential of prostanoid receptors as a treatment target for metabolic disorders.


Assuntos
Prostaglandinas , Receptores de Prostaglandina , Adipócitos/metabolismo , Animais , Feminino , Inflamação/metabolismo , Lipólise , Camundongos , Prostaglandinas/fisiologia , Receptores de Prostaglandina/metabolismo
12.
Am J Respir Crit Care Med ; 206(5): 596-607, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35728047

RESUMO

Rationale: Although persistent fibroblast activation is a hallmark of idiopathic pulmonary fibrosis (IPF), mechanisms regulating persistent fibroblast activation in the lungs have not been fully elucidated. Objectives: On the basis of our observation that lung fibroblasts express TBXA2R (thromboxane-prostanoid receptor) during fibrosis, we investigated the role of TBXA2R signaling in fibrotic remodeling. Methods: We identified TBXA2R expression in lungs of patients with IPF and mice and studied primary mouse and human lung fibroblasts to determine the impact of TBXA2R signaling on fibroblast activation. We used TBXA2R-deficient mice and small-molecule inhibitors to investigate TBXA2R signaling in preclinical lung fibrosis models. Measurements and Main Results: TBXA2R expression was upregulated in fibroblasts in the lungs of patients with IPF and in mouse lungs during experimental lung fibrosis. Genetic deletion of TBXA2R, but not inhibition of thromboxane synthase, protected mice from bleomycin-induced lung fibrosis, thereby suggesting that an alternative ligand activates profibrotic TBXA2R signaling. In contrast to thromboxane, F2-isoprostanes, which are nonenzymatic products of arachidonic acid induced by reactive oxygen species, were persistently elevated during fibrosis. F2-isoprostanes induced TBXA2R signaling in fibroblasts and mediated a myofibroblast activation profile due, at least in part, to potentiation of TGF-ß (transforming growth factor-ß) signaling. In vivo treatment with the TBXA2R antagonist ifetroban reduced profibrotic signaling in the lungs, protected mice from lung fibrosis in three preclinical models (bleomycin, Hermansky-Pudlak mice, and radiation-induced fibrosis), and markedly enhanced fibrotic resolution after bleomycin treatment. Conclusions: TBXA2R links oxidative stress to fibroblast activation during lung fibrosis. TBXA2R antagonists could have utility in treating pulmonary fibrosis.


Assuntos
Fibrose Pulmonar Idiopática , Receptores de Tromboxanos , Animais , Bleomicina/farmacologia , F2-Isoprostanos/metabolismo , Fibroblastos/metabolismo , Humanos , Fibrose Pulmonar Idiopática/genética , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Prostaglandinas/metabolismo , Receptores de Tromboxanos/metabolismo , Tromboxanos/metabolismo , Fator de Crescimento Transformador beta/metabolismo
13.
Biol Pharm Bull ; 45(6): 684-690, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35650096

RESUMO

Since the discovery of ß-arrestin, a new concept/viewpoint has arisen in G-protein coupled receptor (GPCR)-mediated signaling. The Lock and Key concept of GPCR was previously recognized as basically a single- or mono-originated pathway activated from a single receptor. However, the new concept/viewpoint allows for many- or more-than-one-originated pathways activated from a single receptor; namely, biased activities. It is well-recognized that prostanoids exhibit preferences for their corresponding cognate receptors, while promiscuous cross-reactivities have also been reported among endogenous prostanoids and their receptor family. However, of particular interest, such cross-reactivities have led to reports of their physiologically significant roles. Thus, this review discusses and considers that the endogenous prostanoids are not showing random cross-reactivities but what are showing important physiological and pathological activities as biased ligands. Moreover, why and how the biased activities are evoked by endogenous structurally similar prostanoid ligands are discussed. Furthermore, when the biased activities of endogenous prostanoids first arose is also discussed and considered. These biased activities of endogenous prostanoids are also discussed from the perspective that they may provide many benefits and/or disadvantages for all living things, any-where on this planet, who/which are utilizing, had utilized, and will utilize the prostanoids and their receptor system, as a marked driving force for evolution.


Assuntos
Prostaglandinas , Receptores Acoplados a Proteínas G , Ligantes , Receptores Acoplados a Proteínas G/metabolismo
14.
FASEB J ; 36(5): e22293, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35349198

RESUMO

The F prostanoid receptor (FP), which accounts for the therapeutic effect of PGF2α in uterine atony that leads to postpartum hemorrhage and maternal morbidity, could possibly mediate vasoconstrictor effect in small or resistance arteries to elevate blood pressure that limits the clinical use of the agent in patients with cardiovascular disorders. This study aimed to test the above hypothesis with genetically altered mice. Ex vivo and in vivo experiments were performed on control wild-type (WT) mice and mice with deficiencies in FP (FP-/- ) or thromboxane (Tx)-prostanoid receptor (the original receptor of TxA2 ; TP-/- ), and/or those with an additional deficiency in E prostanoid receptor-3 (one of the vasoconstrictor receptors of PGE2 ; EP3-/- ). Here, we show that PGF2α indeed evoked vasoconstrictor responses in the above-mentioned tissues of WT mice, which were however unaltered by FP-/- . Interestingly, such contractile responses were reversed into dilations by TP-/- /EP3-/- . A similar pattern of results was observed with the pressor effect of PGF2α under in vivo conditions. However, TP-/- alone (which could largely remove the contractile responses) did not result in relaxation to PGF2α . Also, either the ex vivo vasodilator effect or the in vivo depressor response of PGF2α obtained after the removal of TP and EP3-mediated actions was unaltered by FP-/- . Therefore, both the ex vivo vasoconstrictor action in small or resistance arteries and the systemic pressor effect of PGF2α can reflect vasoconstrictor activities derived from the non-FP receptors TP and EP3 outweighing a concurrently activated dilator effect, which is again independent of FP.


Assuntos
Receptores de Prostaglandina , Vasoconstritores , Animais , Feminino , Camundongos , Prostaglandinas , Prostaglandinas F , Receptores de Prostaglandina/genética , Receptores de Tromboxanos/genética , Vasoconstritores/farmacologia
15.
Aging Dis ; 13(1): 313-328, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35111376

RESUMO

Aging and hypertension are major risk factors for cerebral small vessel disease (CSVD). Anti-hypertensive therapy has achieved effective; however, incomplete results in treating CSVD, suggesting the need for additional treatments. Targeting abnormal inflammatory responses has become a topic of research interest. Small artery remodeling is the main pathological feature of CSVD. Inhibition of the E-prostanoid 3 (EP3) receptor has been shown to attenuate vascular remodeling in peripheral organs; however, little is known about its role in CSVD. Therefore, we investigated whether the deletion of EP3 attenuates the development of CSVD in an animal model-- stroke-prone renovascular hypertensive rat (RHRsp). We found that the cerebral small arteries of RHRsp exhibited increased EP3 expression. Despite no alleviation of hypertension, the deletion of EP3 still attenuated the cerebral small artery remodeling of RHRsp, as evidenced by reduced overexpression of extracellular matrix (ECM) in the vessel. In vitro experiments indicated that EP3 deletion regulated the expression of ECM by downregulating TGF-ß1/Smad signaling. Furthermore, the Morris water maze test and magnetic resonance test demonstrated that EP3 knockout attenuated cognitive impairment of the RHRsp, possibly through increased cerebral blood flow. Together, our results indicate that the deletion of EP3 attenuates vascular remodeling and vascular cognitive impairment induced by hypertension, and blockade of the EP3 receptor may be a promising strategy for the treatment of CSVD.

16.
Reprod Med Biol ; 21(1): e12423, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34938147

RESUMO

PURPOSE: While the prevailing view holds that the prostaglandin E2 (PGE2) signaling plays a vital role in endometriosis, PGE2 also is known to be anti-fibrotic. We investigated the immunostaining of COX-2, EP2, and EP4, along with fibrotic content in ovarian endometrioma (OE) and deep endometriosis (DE) lesions, and in OE lesions from adolescent and adult patients. In addition, we evaluated the effect of substrate stiffness on the expression of COX-2, EP2, and EP4 in endometrial stromal cells. METHODS: Immunohistochemistry analysis of COX-2, EP2, and EP4, along with the quantification of lesional fibrosis, was conducted for OE and DE lesion samples and also OE lesion samples from adolescent and adult patients. The effect of substrate rigidity on fibroblast-to-myofibroblast transdifferentiation (FMT) and the expression of COX-2, EP2, and EP4, with or without TGF-ß1 stimulation, were investigated. RESULTS: The immunostaining of COX-2, EP2, and EP4 was substantially reduced in endometriotic lesions as lesions became more fibrotic. Both TGF-ß1 stimulation and stiff substrates induced FMT and reduced the expression of COX-2, EP2, and EP4. CONCLUSIONS: Since fibrosis is a common feature of endometriosis, our results thus cast doubts on the use of therapeutics that suppresses the PGE2 signaling pathway, either by inhibiting COX-2 or EP2/EP4.

17.
Reprod Med Biol ; 21(1): e12426, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34938150

RESUMO

PURPOSE: We investigated the change, if any, in prostaglandin E2 (PGE2) signaling in endometriotic lesions of different developmental stages in mouse. In addition, we evaluated the effect of treatment of mice with induced deep endometriosis (DE) with inhibitors of PGE2 receptor subtypes EP2 and EP4 and metformin. METHODS: Three mouse experimentations were conducted. In Experiment 1, female Balb/C mice were induced with endometriosis or DE and were serially sacrificed after induction. Experiments 2 and 3 evaluated the effect of treatment with EP2 and EP4 inhibitors and metformin, respectively, in mice with induced DE. Immunohistochemistry analysis of COX-2, EP2, and EP4, along with the extent of lesional fibrosis, was evaluated. RESULTS: The immunostaining of COX-2, EP2, and EP4 turned from activation to a stall as lesions progressed. Treatment with EP2/EP4 inhibitors in DE mice exacerbated endometriosis-associated hyperalgesia and promoted fibrogenesis in lesions even though it suppressed the PGE2 signaling dose-dependently. In contrast, treatment with metformin resulted in increased PGE2 signaling, concomitant with improved hyperalgesia, and retarded lesional fibrogenesis. CONCLUSIONS: The PGE2 signaling diminishes as endometriotic lesions progress. Treatment with EP2/EP4 inhibitors in DE mice exacerbates endometriosis, but metformin appears to be promising seemingly through the induction of the PGE2 signaling.

18.
Acta Pharm Sin B ; 11(9): 2749-2767, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34589395

RESUMO

Diabetic nephropathy (DN) has been recognized as a severe complication of diabetes mellitus and a dominant pathogeny of end-stage kidney disease, which causes serious health problems and great financial burden to human society worldwide. Conventional strategies, such as renin-angiotensin-aldosterone system blockade, blood glucose level control, and bodyweight reduction, may not achieve satisfactory outcomes in many clinical practices for DN management. Notably, due to the multi-target function, Chinese medicine possesses promising clinical benefits as primary or alternative therapies for DN treatment. Increasing studies have emphasized identifying bioactive compounds and molecular mechanisms of reno-protective effects of Chinese medicines. Signaling pathways involved in glucose/lipid metabolism regulation, antioxidation, anti-inflammation, anti-fibrosis, and podocyte protection have been identified as crucial mechanisms of action. Herein, we summarize the clinical efficacies of Chinese medicines and their bioactive components in treating and managing DN after reviewing the results demonstrated in clinical trials, systematic reviews, and meta-analyses, with a thorough discussion on the relative underlying mechanisms and molecular targets reported in animal and cellular experiments. We aim to provide comprehensive insights into the protective effects of Chinese medicines against DN.

19.
Am J Physiol Renal Physiol ; 320(4): F537-F547, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33491563

RESUMO

Local or systemic inflammation can severely impair urinary bladder functions and contribute to the development of voiding disorders in millions of people worldwide. Isoprostanes are inflammatory lipid mediators that are upregulated in the blood and urine by oxidative stress and may potentially induce detrusor overactivity. The aim of the present study was to investigate the effects and signal transduction of isoprostanes in human and murine urinary bladders in order to provide potential pharmacological targets in detrusor overactivity. Contraction force was measured with a myograph in murine and human urinary bladder smooth muscle (UBSM) ex vivo. Isoprostane 8-iso-PGE2 and 8-iso-PGF2α evoked dose-dependent contraction in the murine UBSM, which was abolished in mice deficient in the thromboxane prostanoid (TP) receptor. The responses remained unaltered after removal of the mucosa or incubation with tetrodotoxin. Smooth muscle-specific deletion of Gα12/13 protein or inhibition of Rho kinase by Y-27632 decreased the contractions. In Gαq/11-knockout mice, responses were reduced and in the presence of Y-27632 abolished completely. In human UBSM, the TP agonist U-46619 evoked dose-dependent contractions. Neither atropine nor the purinergic receptor antagonist pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid decreased the effect, indicating that TP receptors directly mediate detrusor muscle contraction. 8-iso-PGE2 and 8-iso-PGF2α evoked dose-dependent contraction in the human UBSM, and these responses were abolished by the TP antagonist SQ-29548 and were decreased by Y-27632. Our results indicate that isoprostanes evoke contraction in murine and human urinary bladders, an effect mediated by the TP receptor. The G12/13-Rho-Rho kinase pathway plays a significant role in mediating the contraction and therefore may be a potential therapeutic target in detrusor overactivity.NEW & NOTEWORTHY Voiding disorders affect millions of people worldwide. Inflammation can impair urinary bladder functions and contribute to the development of detrusor overactivity. The effects and signal transduction of inflammatory lipid mediator isoprostanes were studied in human and murine urinary bladders ex vivo. We found that isoprostanes evoke contraction, an effect mediated by thromboxane prostanoid receptors. The G12/13-Rho-Rho kinase signaling pathway plays a significant role in mediating the contraction and therefore may be a potential therapeutic target.


Assuntos
Isoprostanos/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Antagonistas de Prostaglandina/farmacologia , Receptores de Prostaglandina/efeitos dos fármacos , Receptores de Tromboxanos/efeitos dos fármacos , Animais , Humanos , Prostaglandinas/farmacologia , Receptores de Tromboxanos/fisiologia
20.
J Matern Fetal Neonatal Med ; 34(1): 41-48, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30810415

RESUMO

Purpose: Mifepristone in conjunction with misoprostol, is widely used in China as an effective medical abortifacient. However, a small proportion of women experience the unpleasant side effects of prolonged vaginal bleeding caused by delayed embryo expulsion. The aims of this study were to determine whether the expression levels of prostanoid receptors in human decidua are associated with delayed embryo expulsion in mifepristone-misoprostol induced an early medical abortion.Methods: Discharged decidua tissues were collected from females undergoing an artificial abortion (AA) (n = 28), females with early embryo expulsion during a medical abortion (EEMA) (n = 20) and delayed embryo expulsion in medical abortion (DEMA) (n = 30). The expression levels of prostanoid receptors in human decidua were assessed with immunohistochemistry and real-time PCR methods. Further, the RNAi method was used to silence prostanoid receptors 4 (EP4) in the primary decidual cells and human endometrial adenocarcinoma cell line Ishikawa cells in vitro and cell cycle analysis of these cells was performed.Results: All five prostanoid receptors (EP1-4, FP) were observed in human early pregnancy decidua. The protein and mRNA expression level of EP4 in the DEMA group were all significantly higher than that in the EEMA group. EP4 silence induced G1/S arrest of primary decidual cells and Ishikawa cells in vitro.Conclusions: Elevated expression level of EP4 in human decidua was significantly associated with delayed embryo expulsion in early medical abortion by promoting decidual cell proliferation. Detailed studies on the nature of roles EP4 plays in human decidua will help us to develop more effective prevention and noninvasive intervention approaches for delayed embryo expulsion during a medical abortion.


Assuntos
Aborto Induzido , Misoprostol , Proliferação de Células , China , Decídua , Feminino , Humanos , Mifepristona/farmacologia , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA