RESUMEN
Regulatory T cells (Treg cells) are important for preventing autoimmunity and maintaining tissue homeostasis, but whether Treg cells can adopt tissue- or immune-context-specific suppressive mechanisms is unclear. Here, we found that the enzyme hydroxyprostaglandin dehydrogenase (HPGD), which catabolizes prostaglandin E2 (PGE2) into the metabolite 15-keto PGE2, was highly expressed in Treg cells, particularly those in visceral adipose tissue (VAT). Nuclear receptor peroxisome proliferator-activated receptor-γ (PPARγ)-induced HPGD expression in VAT Treg cells, and consequential Treg-cell-mediated generation of 15-keto PGE2 suppressed conventional T cell activation and proliferation. Conditional deletion of Hpgd in mouse Treg cells resulted in the accumulation of functionally impaired Treg cells specifically in VAT, causing local inflammation and systemic insulin resistance. Consistent with this mechanism, humans with type 2 diabetes showed decreased HPGD expression in Treg cells. These data indicate that HPGD-mediated suppression is a tissue- and context-dependent suppressive mechanism used by Treg cells to maintain adipose tissue homeostasis.
Asunto(s)
Dinoprostona/análogos & derivados , Dinoprostona/metabolismo , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Grasa Intraabdominal/inmunología , Linfocitos T Reguladores/enzimología , Linfocitos T Reguladores/inmunología , Células 3T3 , Animales , Línea Celular , Diabetes Mellitus Tipo 2/metabolismo , Células HEK293 , Homeostasis/inmunología , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Resistencia a la Insulina/genética , Grasa Intraabdominal/citología , Células Jurkat , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Noqueados , Factor de Transcripción STAT5/metabolismoRESUMEN
BACKGROUND AIMS: Cholangiocarcinoma (CCA) is a highly lethal malignancy originating from the biliary ducts. Current CCA diagnostic and prognostic assessments cannot satisfy the clinical requirement. Bile detection is rarely performed, and herein, we aim to estimate the clinical significance of bile liquid biopsy by assessing bile exosomal concentrations and components. APPROACH RESULTS: Exosomes in bile and sera from CCA, pancreatic cancer, and common bile duct stone were identified and quantified by transmission electronmicroscopy, nanoparticle tracking analysis, and nanoFCM. Exosomal components were assessed by liquid chromatography with tandem mass spectrometry and microRNA sequencing (miRNA-seq). Bile exosomal concentration in different diseases had no significant difference, but miR-182-5p and miR-183-5p were ectopically upregulated in CCA bile exosomes. High miR-182/183-5p in both CCA tissues and bile indicates a poor prognosis. Bile exosomal miR-182/183-5p is secreted by CCA cells and can be absorbed by biliary epithelium or CCA cells. With xenografts in humanized mice, we showed that bile exosomal miR-182/183-5p promotes CCA proliferation, invasion, and epithelial-mesenchymal transition (EMT) by targeting hydroxyprostaglandin dehydrogenase in CCA cells and mast cells (MCs), and increasing prostaglandin E2 generation, which stimulates PTGER1 and increases CCA stemness. In single-cell mRNA-seq, hydroxyprostaglandin dehydrogenase is predominantly expressed in MCs. miR-182/183-5p prompts MC to release VEGF-A release from MC by increasing VEGF-A expression, which facilitates angiogenesis. CONCLUSIONS: CCA cells secret exosomal miR-182/183-5p into bile, which targets hydroxyprostaglandin dehydrogenase in CCA cells and MCs and increases prostaglandin E2 and VEGF-A release. Prostaglandin E2 promotes stemness by activating PTGER1. Our results reveal a type of CCA self-driven progression dependent on bile exosomal miR-182/183-5p and MCs, which is a new interplay pattern of CCA and bile.
Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , MicroARNs , Humanos , Animales , Ratones , Dinoprostona , MicroARNs/genética , Bilis/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Neoplasias de los Conductos Biliares/patología , Línea Celular Tumoral , Colangiocarcinoma/patología , Conductos Biliares Intrahepáticos/patología , Hidroxiprostaglandina Deshidrogenasas/genética , Proliferación Celular , Regulación Neoplásica de la Expresión GénicaRESUMEN
Non-steroidal anti-inflammatory drugs (NSAIDs), the most highly prescribed drugs in the world for the treatment of pain, inflammation, and fever, cause gastric mucosal damage, including ulcers, directly or indirectly, by which the development of GI-safer (-sparing) NSAIDs relates to unmet medical needs. This study aimed to document the preventive effects of walnut polyphenol extracts (WPEs) against NSAID-induced gastric damage along with the molecular mechanisms. RGM-1 gastric mucosal cells were administered with indomethacin, and the expressions of the inflammatory mediators between indomethacin alone or a combination with WPEs were compared. The expressions of the inflammatory mediators, including COX-1 and COX-2, prostaglandin E2, 15-hydroxyprostaglandin dehydrogenase (15-PGDH), and antioxidant capacity, were analyzed by Western blot analysis, RT-PCR, and ELISA, respectively. HO-1, Nrf-2, and keap1 were investigated. The in vivo animal models were followed with in vitro investigations. The NSAIDs increased the expression of COX-2 and decreased COX-1 and 15-PGDH, but the WPEs significantly attenuated the NSAID-induced COX-2 expression. Interestingly, the WPEs induced the expression of 15-PGDH. By using the deletion constructs of the 15-PGDH promoter, we found that c-Jun is the most essential determinant of the WPE-induced up-regulation of 15-PGDH expression. We confirmed that the knockdown of c-Jun abolished the ability of the WPEs to up-regulate the 15-PGDH expression. In addition, the WPEs significantly increased the HO-1 expression. The WPEs increased the nuclear translocation of Nrf2 by Keap-1 degradation, and silencing Nrf2 markedly reduced the WPE-induced HO-1 expression. We found that the WPE-induced HO-1 up-regulation was attenuated in the cells harboring the mutant Keap1, in which the cysteine 151 residue was replaced by serine. These in vitro findings were exactly validated in indomethacin-induced gastric rat models. Daily walnut intake can be a promising nutritional supplement providing potent anti-inflammatory, antioxidative, and mucosa-protective effects against NSAID-induced GI damage.
Asunto(s)
Mucosa Gástrica , Hidroxiprostaglandina Deshidrogenasas , Indometacina , Juglans , Factor 2 Relacionado con NF-E2 , Animales , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Indometacina/efectos adversos , Juglans/química , Ratas , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Hidroxiprostaglandina Deshidrogenasas/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/genética , Masculino , Extractos Vegetales/farmacología , Antiinflamatorios no Esteroideos/farmacología , Hemo-Oxigenasa 1/metabolismo , Hemo-Oxigenasa 1/genética , Línea Celular , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Polifenoles/farmacologíaRESUMEN
Increased androgen receptor (AR) signaling brought on by higher intratumoral androgen production and AR amplification is associated with castrate-resistant prostate cancer (CRPC). Cell proliferation in this case continues even during low expression of testosterone in the body. Aldo-keto reductase family 1 member C3 (AKR1C3) is one of the most elevated genes in CRPC and catalyzes the formation of powerful AR ligands from inactive forms. The current work aimed to use the x-ray method to investigate the ligand's crystal structure while also conducting molecular docking and molecular dynamics tests on the synthesized molecules against AKR1C3. As per the results obtained, the MM-PBSA binding energies of inhibitors 2,2'-((4-methoxyphenyl)methylene)bis(3,4-hydroxy-5,5-dimethylcyclohex-2-en-1-one is -132.456 kJ mol-1 and 2,2'-(phenylmethylene)bis(3-hydroxy-5,5-dimethylcyclohex-2-en-1-one is -81.017 kJ mol-1 . These results create a promising approach to drug design based on its fit to the structures of the receptor site rather than basing it on analogies to other active structures.
Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Masculino , Humanos , Simulación del Acoplamiento Molecular , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Hidroxiprostaglandina Deshidrogenasas/genética , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Simulación de Dinámica Molecular , Línea Celular TumoralRESUMEN
The arachidonic acid cascade is a major inflammatory pathway that produces prostaglandin E2 (PGE2). Although inhibition of 15-hydroxyprostaglandin dehydrogenase (15-PGDH) is reported to lead to PGE2 accumulation, the role of 15-PGDH expression in the tumor microenvironment remains unclear. We utilized Panc02 murine pancreatic cancer cells for orthotopic transplantation into wild-type and 15-pgdh+/- mice and found that 15-pgdh depletion in the tumor microenvironment leads to enhanced tumorigenesis accompanied by an increase in cancer-associated fibroblasts (CAFs) and the promotion of fibrosis. The fibrotic tumor microenvironment is widely considered to be hypovascular; however, we found that the angiogenesis level is maintained in 15-pgdh+/- mice, and these changes were also observed in a genetically engineered PDAC mouse model. Further confirmation revealed that fibroblast growth factor 1 (FGF1) is secreted by pancreatic cancer cells after PGE2 stimulation, consequently promoting CAF proliferation and vascular endothelial growth factor A (VEGFA) expression in the tumor microenvironment. Finally, in 15-pgdh+/- Acta2-TK mice, depletion of fibroblasts inhibited angiogenesis and cancer cell viability in orthotopically transplanted tumors. These findings highlighted the role of 15-pgdh downregulation in enhancing PGE2 accumulation in the pancreatic tumor microenvironment and in subsequently maintaining the angiogenesis level in fibrotic tumors along with CAF expansion.
Asunto(s)
Neoplasias Pancreáticas , Factor A de Crecimiento Endotelial Vascular , Animales , Ácido Araquidónico , Línea Celular Tumoral , Dinoprostona/metabolismo , Dinoprostona/farmacología , Factor 1 de Crecimiento de Fibroblastos , Fibrosis , Hidroxiprostaglandina Deshidrogenasas/genética , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Ratones , Neoplasias Pancreáticas/genética , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/genética , Neoplasias PancreáticasRESUMEN
BACKGROUND AND AIMS: Alcohol-associated liver disease (ALD) is a common chronic liver disease worldwide with high morbidity and mortality, and no Food and Drug Administration-approved therapies. Fructose (dietary or endogenous), its metabolite uric acid, and aldose reductase (AR, the only endogenous enzyme that produces fructose) are strongly associated with the development of nonalcoholic fatty liver disease. However, the role of AR or its metabolites in ALD remains understudied and was examined using human specimens, cultured cells, and mouse model systems. APPROACH AND RESULTS: We demonstrated in liver specimens from patients with alcoholic hepatitis, the AR up-regulation and elevated AR metabolites (sorbitol, fructose, and uric acid), which correlated significantly with (1) increased lipid peroxidation byproducts and endoplasmic reticulum (ER) stress, (2) decreased protective ER chaperones, and (3) greater cell death and liver injury. Furthermore, we established a causal role for AR in ALD by showing that the genetic deficiency of AR (knockout mice) prevented alcohol-induced increase in harmful AR metabolites, toxic aldehydes, steatosis, ER stress, apoptosis, and liver injury. Finally, we demonstrated the therapeutic potential of pharmacological AR inhibition against alcohol-induced hepatic injury in experimental ALD. CONCLUSIONS: Our data demonstrate that hepatic AR up-regulation, and consequent elevation in fructose, sorbitol and/or uric acid, are important factors contributing to alcohol-induced steatosis, ER stress, apoptosis, and liver injury in both experimental and human ALD. Our study provides a strong rationale to evaluate AR as a potential therapeutic target and to test AR inhibitors to ameliorate alcohol-induced liver injury.
Asunto(s)
Aldehído Reductasa/metabolismo , Fructosa/sangre , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Hepatopatías Alcohólicas/metabolismo , Ácido Úrico/sangre , Adulto , Aldehído Reductasa/genética , Animales , Apoptosis/efectos de los fármacos , Estudios de Casos y Controles , Estudios de Cohortes , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Etanol/administración & dosificación , Etanol/toxicidad , Femenino , Fructosa/metabolismo , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Hígado/efectos de los fármacos , Hígado/patología , Hepatopatías Alcohólicas/sangre , Hepatopatías Alcohólicas/diagnóstico , Hepatopatías Alcohólicas/etiología , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Estrés Oxidativo/efectos de los fármacos , Índice de Severidad de la Enfermedad , Sorbitol/sangre , Sorbitol/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Ácido Úrico/metabolismoRESUMEN
Rat genes, akr1c19 and RGD1564865, encode members (R1C19 and 20HSDL, respectively) of the aldo-keto reductase (AKR) 1C subfamily, whose functions, however, remain unknown. Here, we show that recombinant R1C19 and 20HSDL exhibit NAD+-dependent dehydrogenase activity for prostaglandins (PGs) with 9α-hydroxy group (PGF2α, its 13,14-dihydro- and 15-keto derivatives, 9α,11ß-PGF2 and PGD2). 20HSDL oxidized the PGs with much lower Km (0.3-14 µM) and higher kcat/Km values (0.064-2.6 min-1µM-1) than those of R1C19. They also differed in other properties: R1C19, but not 20HSDL, oxidized some 17ß-hydroxysteroids (5ß-androstane-3α,17ß-diol and 5ß-androstan-17ß-ol-3-one). 20HSDL was specifically inhibited by zomepirac, but not by R1C19-selective inhibitors (hexestrol, flavonoids, ibuprofen and flufenamic acid), although the two enzymes were sensitive to indomethacin and cis-unsaturated fatty acids. The mRNA for 20HSDL was expressed abundantly in rat kidney and at low levels in the liver, testis, brain, heart and colon, in contrast to ubiquitous expression of R1C19 mRNA. The comparison of enzymic features of R1C19 and 20HSDL with rat PG dehydrogenases and other AKRs suggests not only a close relationship of 20HSDL with 9-hydroxy-PG dehydrogenase in rat kidney, but also roles of R1C19 and rat AKRs (1C16 and 1C24) in the metabolism of PGF2α, PGD2 and 9α,11ß-PGF2 in other tissues.
Asunto(s)
Aldo-Ceto Reductasas/biosíntesis , Regulación Enzimológica de la Expresión Génica , Hidroxiprostaglandina Deshidrogenasas/biosíntesis , Hidroxiesteroides/metabolismo , Aldo-Ceto Reductasas/genética , Animales , Hidroxiprostaglandina Deshidrogenasas/genética , Especificidad de Órganos , Oxidación-Reducción , RatasRESUMEN
Eicosanoids comprise a diverse group of bioactive lipids which orchestrate inflammation, immunity, and tissue homeostasis, and whose dysregulation has been implicated in carcinogenesis. Among the various eicosanoid metabolic pathways, studies of their role in endometrial cancer (EC) have very much been confined to the COX-2 pathway. This study aimed to determine changes in epithelial eicosanoid metabolic gene expression in endometrial carcinogenesis; to integrate these with eicosanoid profiles in matched clinical specimens; and, finally, to investigate the prognostic value of candidate eicosanoid metabolic enzymes. Eicosanoids and related mediators were profiled using liquid chromatography-tandem mass spectrometry in fresh frozen normal, hyperplastic, and cancerous (types I and II) endometrial specimens (n = 192). Sample-matched epithelia were isolated by laser capture microdissection and whole genome expression analysis was performed using microarrays. Integration of eicosanoid and gene expression data showed that the accepted paradigm of increased COX-2-mediated prostaglandin production does not apply in EC carcinogenesis. Instead, there was evidence for decreased PGE2 /PGF2α inactivation via 15-hydroxyprostaglandin dehydrogenase (HPGD) in type II ECs. Increased expression of 5-lipoxygenase (ALOX5) mRNA was also identified in type II ECs, together with proportional increases in its product, 5-hydroxyeicosatetraenoic acid (5-HETE). Decreased HPGD and elevated ALOX5 mRNA expression were associated with adverse outcome, which was confirmed by immunohistochemical tissue microarray analysis of an independent series of EC specimens (n = 419). While neither COX-1 nor COX-2 protein expression had prognostic value, low HPGD combined with high ALOX5 expression was associated with the worst overall and progression-free survival. These findings highlight HPGD and ALOX5 as potential therapeutic targets in aggressive EC subtypes. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Asunto(s)
Araquidonato 5-Lipooxigenasa/metabolismo , Carcinoma Endometrioide/enzimología , Eicosanoides/metabolismo , Neoplasias Endometriales/enzimología , Células Epiteliales/enzimología , Adulto , Anciano , Anciano de 80 o más Años , Araquidonato 5-Lipooxigenasa/genética , Carcinoma Endometrioide/genética , Carcinoma Endometrioide/patología , Carcinoma Endometrioide/terapia , Cromatografía Líquida de Alta Presión , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Neoplasias Endometriales/terapia , Células Epiteliales/patología , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Metabolómica/métodos , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Supervivencia sin Progresión , Estudios Prospectivos , Espectrometría de Masas en Tándem , Regulación hacia ArribaRESUMEN
In recent years, it has been established that programmed cell death protein ligand 1 (PD-L1)-mediated inhibition of activated PD-1+ T lymphocytes plays a major role in tumor escape from immune system during cancer progression. Lately, the anti-PD-L1 and -PD-1 immune therapies have become an important tool for treatment of advanced human cancers, including bladder cancer. However, the underlying mechanisms of PD-L1 expression in cancer are not fully understood. We found that coculture of murine bone marrow cells with bladder tumor cells promoted strong expression of PD-L1 in bone marrow-derived myeloid cells. Tumor-induced expression of PD-L1 was limited to F4/80+ macrophages and Ly-6C+ myeloid-derived suppressor cells. These PD-L1-expressing cells were immunosuppressive and were capable of eliminating CD8 T cells in vitro. Tumor-infiltrating PD-L1+ cells isolated from tumor-bearing mice also exerted morphology of tumor-associated macrophages and expressed high levels of prostaglandin E2 (PGE2)-forming enzymes microsomal PGE2 synthase 1 (mPGES1) and COX2. Inhibition of PGE2 formation, using pharmacologic mPGES1 and COX2 inhibitors or genetic overexpression of PGE2-degrading enzyme 15-hydroxyprostaglandin dehydrogenase (15-PGDH), resulted in reduced PD-L1 expression. Together, our study demonstrates that the COX2/mPGES1/PGE2 pathway involved in the regulation of PD-L1 expression in tumor-infiltrating myeloid cells and, therefore, reprogramming of PGE2 metabolism in tumor microenvironment provides an opportunity to reduce immune suppression in tumor host.
Asunto(s)
Antígeno B7-H1/biosíntesis , Células de la Médula Ósea/metabolismo , Ciclooxigenasa 2/fisiología , Dinoprostona/fisiología , Macrófagos/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Prostaglandina-E Sintasas/fisiología , Animales , Antígeno B7-H1/genética , Comunicación Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Humanos , Hidroxiprostaglandina Deshidrogenasas/biosíntesis , Hidroxiprostaglandina Deshidrogenasas/genética , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos NOD , Ratones SCID , Antagonistas de Prostaglandina/farmacología , Neoplasias de la Vejiga Urinaria/inmunología , Neoplasias de la Vejiga Urinaria/patologíaRESUMEN
We previously reported that single cells from a human colorectal cancer (CRC) cell line (HCA-7) formed either hollow single-layered polarized cysts or solid spiky masses when plated in 3D in type-I collagen. To begin in-depth analyses into whether clonal cysts and spiky masses possessed divergent properties, individual colonies of each morphology were isolated and expanded. The lines thus derived faithfully retained their parental cystic and spiky morphologies and were termed CC (cystic) and SC (spiky), respectively. Although both CC and SC expressed EGF receptor (EGFR), the EGFR-neutralizing monoclonal antibody, cetuximab, strongly inhibited growth of CC, whereas SC was resistant to growth inhibition, and this was coupled to increased tyrosine phosphorylation of MET and RON. Addition of the dual MET/RON tyrosine kinase inhibitor, crizotinib, restored cetuximab sensitivity in SC. To further characterize these two lines, we performed comprehensive genomic and transcriptomic analysis of CC and SC in 3D. One of the most up-regulated genes in CC was the tumor suppressor 15-PGDH/HPGD, and the most up-regulated gene in SC was versican (VCAN) in 3D and xenografts. Analysis of a CRC tissue microarray showed that epithelial, but not stromal, VCAN staining strongly correlated with reduced survival, and combined epithelial VCAN and absent HPGD staining portended a poorer prognosis. Thus, with this 3D system, we have identified a mode of cetuximab resistance and a potential prognostic marker in CRC. As such, this represents a potentially powerful system to identify additional therapeutic strategies and disease-relevant genes in CRC and possibly other solid tumors.
Asunto(s)
Técnicas de Cultivo de Célula/métodos , Cetuximab/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Resistencia a Antineoplásicos/genética , Animales , Antineoplásicos Inmunológicos/farmacología , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Crizotinib , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Ratones , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazoles/farmacología , Piridinas/farmacología , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Análisis de Matrices Tisulares , Versicanos/genética , Ensayos Antitumor por Modelo de XenoinjertoRESUMEN
Gastric cancer (GC) represents the third leading cause of cancer-related deaths worldwide. The levels of prostaglandin E2, a key player in the hallmarks of cancer, are mainly regulated by prostaglandin-endoperoxide synthase 2 (PTGS2) and ATP-binding cassette subfamily C member 4 (ABCC4), involved in its synthesis and exportation, respectively, and 15-hydroxyprostaglandin dehydrogenase (15-PGDH) and solute carrier organic anion transporter family member 2A1 (SLCO2A1), responsible for its inactivation. Even though there are distinct molecular signatures across ethnic populations, most published studies focus on Asian populations. Our main aim was to explore the genetic expression of the aforementioned molecules in a Caucasian population. 94 "Normal" and 89 tumoral formalin-fixed paraffin-embedded (FFPE) samples from GC patients were used to assess the mRNA expression of PTGS2, ABCC4, hydroxyprostaglandin dehydrogenase 15-(NAD) (HPGD), SLCO2A1 by Real-Time PCR. We found an upregulation for the PTGS2 gene mean factor of 2.51 and a downregulation for the HPGD and SLCO2A1 genes (mean factor of 0.10 and 0.37, respectively) in tumorous mucosa in a gender-independent manner. In females, we observed an ABCC4 downregulation and a PTGS2 mRNA upregulation compared to males in tumoral mucosa (mean factor of 0.61 and 1.64, respectively). We reported dysregulation of the inflammation triggered PGE2 pathway in a Caucasian population with an intermediate risk for GC, which might highlight the applicability of aspirin in the treatment of GC patients.
Asunto(s)
Adenocarcinoma/metabolismo , Dinoprostona/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Gástricas/metabolismo , Adenocarcinoma/genética , Anciano , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Dinoprostona/genética , Femenino , Mucosa Gástrica/metabolismo , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Masculino , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Neoplasias Gástricas/genéticaRESUMEN
Chorionic NAD-dependent 15-hydroxyprostaglandin dehydrogenase (PGDH) plays a pivotal role in controlling the amount of prostaglandins in the uterus and has been implicated in the process of labor. Prior studies identified hydrogen sulfide-generating enzymes cystathionine-ß-synthetase (CBS) and cystathionine-γ-lyase (CSE) in fetal membranes. We investigated whether hydrogen sulfide is involved in the regulation of PGDH expression in the chorion during labor. The chorionic tissues were obtained from pregnant women at preterm in labor and at term in labor or not in labor at term. Levels of CSE and CBS and hydrogen sulfide production rate were down-regulated in term in labor and preterm in labor groups compared with not in labor at term group. The CBS level correlated to PGDH expression in the chorion. Hydrogen sulfide donor NaHS and precursor l-cysteine dose-dependently stimulated PGDH expression and activity in cultured chorionic trophoblasts. The effect of l-cysteine was blocked by CBS inhibitor and CBS siRNA but not by CSE inhibitor and CSE siRNA. Hydrogen sulfide treatment suppressed miR-26b and miR-199a expression in chorionic trophoblasts. miR-26b and miR-199a mimics blocked hydrogen sulfide upregulation of PGDH expression. Our results indicate that hydrogen sulfide plays pivotal roles in maintenance of PGDH expression in the chorion during human pregnancy. Reduced expression of hydrogen sulfide-generating enzymes contributes to an increased amount of prostaglandins in the uterus during labor.
Asunto(s)
Corion/enzimología , Cistationina betasintasa/metabolismo , Cistationina gamma-Liasa/metabolismo , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Trabajo de Parto Prematuro/metabolismo , Nacimiento a Término/metabolismo , Cistationina gamma-Liasa/genética , Regulación hacia Abajo , Femenino , Humanos , Sulfuro de Hidrógeno/metabolismo , Hidroxiprostaglandina Deshidrogenasas/genética , Trabajo de Parto Prematuro/genética , Embarazo , Nacimiento a Término/genéticaRESUMEN
Prostaglandins (PGs) and prostaglandin endoperoxide synthase (PTGS) are considered to be relevant for spermatogenesis and steroidogenesis. PTGS2, prostaglandin F synthase (PGFS) and PGF receptor (PTGFR) are investigated in the adult male dog using the model of the GnRH-agonist implant downregulated canine testis and its subsequent restart of spermatogenesis following abolition of treatment (3, 6, 9, 12 weeks after implant removal). On the mRNA level (ratio), PTGS2, PGFS and PTGFR expression did not differ between downregulation, different stages of recovery of spermatogenesis and untreated adult controls (CG). On the protein level, Sertoli and Leydig cells in all samples and some peritubular cells stained immunopositive for PTGS2. In the tubular compartment, the percentage of the PTGS2-immunopositive area (PIA) and the mean PTGS2-staining intensity (gray scale, GS) did not differ between groups but in the interstitial compartment, the PIA (p = 0.0494) and the GS (p < 0.0001) were significantly upregulated during early recrudescence (week 3/6). Comparing downregulation by two GnRH-agonist implants with juvenile controls (JG) and CG, the mRNA expression (ratio) did not differ. In the tubular compartment, the GS (p = 0.0321) was significantly higher at downregulation compared to CG and in the interstitial compartment, the PIA (p = 0.0073) and the GS (p = 0.0097) were significantly higher in JG compared to downregulation/CG. PTGS2, PGFS and PTGFR mRNA and PTGS2 protein are regularly expressed in the adult, juvenile and downregulated canine testis and downregulation and subsequent recrudescence affect PTGS2 protein expression mainly in Leydig cells. PTGS2 expression in the downregulated testis resembles the one in seasonal Syrian hamster but not juvenile canine testis.
Asunto(s)
Ciclooxigenasa 2/metabolismo , Regulación hacia Abajo , Hormona Liberadora de Gonadotropina/agonistas , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Receptores de Prostaglandina/metabolismo , Espermatogénesis , Animales , Ciclooxigenasa 2/genética , Perros , Regulación hacia Abajo/genética , Hidroxiprostaglandina Deshidrogenasas/genética , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Prostaglandina/genética , Espermatogénesis/genéticaRESUMEN
Endometrial cancer is one of the most common cancers in women worldwide, affecting more than 300,000 women annually. Dysregulated gene expression, especially those mediated by microRNAs, play important role in the development and progression of cancer. This study aimed to investigate differentially expressed genes in endometrial adenocarcinoma using next generation sequencing (NGS) and bioinformatics. The gene expression profiles and microRNA profiles of endometrial adenocarcinoma (cancer part) and normal endometrial tissue (non-cancer part) were assessed with NGS. We identified 56 significantly dysregulated genes, including 47 upregulated and 9 downregulated genes, in endometrial adenocarcinoma. Most of these genes were associated with defense response, response to stimulus, and immune system process, and further pathway analysis showed that human papillomavirus infection was the most significant pathway in endometrial adenocarcinoma. In addition, these genes were also associated with decreased cell death and survival as well as increased cellular movement. The analyses using Human Protein Atlas, identified 6 genes (PEG10, CLDN1, ASS1, WNT7A, GLDC, and RSAD2) significantly associated with poorer prognosis and 3 genes (SFN, PIGR, and CDKN1A) significantly associated with better prognosis. Combining with the data of microRNA profiles using microRNA target predicting tools, two significantly dysregulated microRNA-mediated gene expression changes in endometrial adenocarcinoma were identified: downregulated hsa-miR-127-5p with upregulated CSTB and upregulated hsa-miR-218-5p with downregulated HPGD. These findings may contribute important new insights into possible novel diagnostic or therapeutic strategies for endometrial adenocarcinoma.
Asunto(s)
Adenocarcinoma/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Endometriales/genética , Regulación Neoplásica de la Expresión Génica , Adenocarcinoma/inmunología , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Anciano , Biomarcadores de Tumor/genética , Biología Computacional , Cistatina B/genética , Regulación hacia Abajo , Neoplasias Endometriales/inmunología , Neoplasias Endometriales/mortalidad , Neoplasias Endometriales/patología , Endometrio/patología , Femenino , Estudios de Seguimiento , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Estimación de Kaplan-Meier , MicroARNs/metabolismo , Persona de Mediana Edad , Pronóstico , Transducción de Señal/genética , Transducción de Señal/inmunología , Regulación hacia ArribaRESUMEN
Cyclooxygenases are responsible for the production of prostaglandin H2 (PGH2) from arachidonic acid. PGH2 can be converted into some bioactive prostaglandins, including prostaglandin F2α (PGF2α), a potent chemical messenger used as a biological regulator in the fields of obstetrics and gynecology. The chemical messenger PGF2α has been industrially produced by chemical synthesis. To develop a biotechnological process, in which PGF2α can be produced by a microorganism, we transformed an oleaginous fungus, Mortierella alpina 1S-4, rich in triacylglycerol consisting of arachidonic acid using a cyclooxygenase gene from a red alga, Gracilaria vermiculophylla. PGF2α was accumulated not only in the mycelia of the transformants but also in the extracellular medium. After 12 days of cultivation approximately 860 ng/g and 6421 µg/L of PGF2α were accumulated in mycelia and the extracellular medium, respectively. The results could facilitate the development of novel fermentative methods for the production of prostanoids using an oleaginous fungus.
Asunto(s)
Proteínas Algáceas/genética , Ácido Araquidónico/metabolismo , Dinoprost/biosíntesis , Gracilaria/química , Ingeniería Metabólica/métodos , Mortierella/genética , Prostaglandina-Endoperóxido Sintasas/genética , Agrobacterium tumefaciens/genética , Agrobacterium tumefaciens/metabolismo , Proteínas Algáceas/metabolismo , Medios de Cultivo/química , Expresión Génica , Gracilaria/genética , Hidroxiprostaglandina Deshidrogenasas/genética , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Mortierella/metabolismo , Micelio/genética , Micelio/metabolismo , Plásmidos/química , Plásmidos/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Transformación Genética , TransgenesRESUMEN
Chronic inflammation has a crucial role in cancer development and the progression of various tumors, including pancreatic ductal adenocarcinoma (PDAC). The arachidonate cascade is a major inflammatory pathway that produces several metabolites, such as prostaglandin E2. The enzyme 15-hydroxyprostaglandin dehydrogenase (15-PGDH) degrades prostaglandin and is frequently decreased in several types of cancer; however, the molecular mechanisms of 15-PGDH suppression are unclear. The current study was carried out to elucidate the molecular mechanisms and clinical significance of 15-PGDH suppression in PDAC. Here, we showed that interleukin-1ß (IL-1ß), a pro-inflammatory cytokine, downregulates 15-PGDH expression in PDAC cells, and that IL-1ß expression was inversely correlated with 15-PGDH levels in frozen PDAC tissues. We also found that activated macrophages produced IL-1ß and reduced 15-PGDH expression in PDAC cells. Furthermore, the number of CD163-positive tumor-associated macrophages was shown to be inversely correlated with 15-PGDH levels in PDAC cells by immunohistochemical staining of 107 PDAC samples. Finally, we found that low 15-PGDH expression was significantly associated with advanced tumors, presence of lymph node metastasis and nerve invasion, and poor prognosis in PDAC patients. Our results indicate that IL-1ß derived from TAMs suppresses 15-PGDH expression in PDAC cells, resulting in poor prognosis of PDAC patients.
Asunto(s)
Carcinoma Ductal Pancreático/patología , Regulación hacia Abajo , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Interleucina-1beta/genética , Macrófagos/patología , Masculino , Invasividad Neoplásica , Metástasis de la Neoplasia , Estadificación de Neoplasias , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Pronóstico , Análisis de SupervivenciaRESUMEN
Primary hypertrophic osteoarthropathy (PHO) is a congenital multisystemic entity characterized by three major clinical symptoms: pachydermia, periostosis, and digital clubbing. Recently it has been reported that pathogenic mutations in two genes are known to be associated with PHO: HPGD and SLCO2A1. In the present study, a five-generation consanguineous Pakistani family harboring primary hypertrophic osteoarthropathy in autosomal-recessive pattern was ascertained. Whole genome single nucleotide polymorphisms (SNPs) genotyping and sequence analysis revealed a novel homozygous missense mutation (c.577TËC) of the human HPGD gene in all affected members of the family. The study presented here demonstrate the first case of primary hypertrophic osteoarthropathy reported in Pashtun population.
Asunto(s)
Hidroxiprostaglandina Deshidrogenasas/genética , Mutación Missense , Osteoartropatía Hipertrófica Primaria/genética , Anciano , Niño , Consanguinidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pakistán , Linaje , Polimorfismo de Nucleótido SimpleRESUMEN
BACKGROUND & AIMS: Prostaglandin E2 (PGE2) is mediator of inflammation that regulates tissue regeneration, but its continual activation has been associated with carcinogenesis. Little is known about factors in the PGE2 signaling pathway that contribute to tumor formation. We investigated whether yes-associated protein 1 (YAP1), a transcriptional co-activator in the Hippo signaling pathway, mediates PGE2 function. METHODS: DLD-1 and SW480 colon cancer cell lines were transfected with vectors expressing transgenes or small hairpin RNAs and incubated with recombinant PGE2, with or without pharmacologic inhibitors of signaling proteins, and analyzed by immunoblot, immunofluorescence, quantitative reverse-transcription polymerase chain reaction, transcriptional reporter, and proliferation assays. Dextran sodium sulfate (DSS) was given to induce colitis in C57/BL6 (control) mice, as well as in mice with disruption of the hydroxyprostaglandin dehydrogenase 15 gene (15-PGDH-knockout mice), Yap1 gene (YAP-knockout mice), and double-knockout mice. Some mice also were given indomethacin to block PGE2 synthesis. 15-PGDH knockout mice were crossed with mice with intestine-specific disruption of the salvador family WW domain containing 1 gene (Sav1), which encodes an activator of Hippo signaling. We performed immunohistochemical analyses of colon biopsy samples from 26 patients with colitis-associated cancer and 51 age-and sex-matched patients with colorectal cancer (without colitis). RESULTS: Incubation of colon cancer cell lines with PGE2 led to phosphorylation of cyclic adenosine monophosphate-responsive element binding protein 1 and increased levels of YAP1 messenger RNA, protein, and YAP1 transcriptional activity. This led to increased transcription of the prostaglandin-endoperoxide synthase 2 gene (PTGS2 or cyclooxygenase 2) and prostaglandin E-receptor 4 gene (PTGER4 or EP4). Incubation with PGE2 promoted proliferation of colon cancer cell lines, but not cells with knockdown of YAP1. Control mice developed colitis after administration of DSS, but injection of PGE2 led to colon regeneration in these mice. However, YAP-knockout mice did not regenerate colon tissues and died soon after administration of DSS. 15-PGDH-knockout mice regenerated colon tissues more rapidly than control mice after withdrawal of DSS, and had faster recovery of body weight, colon length, and colitis histology scores. These effects were reversed by injection of indomethacin. SAV1-knockout or 15-PGDH-knockout mice did not develop spontaneous tumors after colitis induction, but SAV1/15-PGDH double-knockout mice developed polyps that eventually progressed to carcinoma in situ. Administration of indomethacin to these mice prevented spontaneous tumor formation. Levels of PGE2 correlated with those of YAP levels in human sporadic colorectal tumors and colitis-associated tumors. CONCLUSIONS: PGE2 signaling increases the expression and transcriptional activities of YAP1, leading to increased expression of cyclooxygenase 2 and EP4 to activate a positive signaling loop. This pathway promotes proliferation of colon cancer cell lines and colon tissue regeneration in mice with colitis. Constitutive activation of this pathway led to formation of polyps and colon tumors in mice.
Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colon/efectos de los fármacos , Neoplasias Colorrectales/genética , Dinoprostona/farmacología , Fosfoproteínas/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , Regeneración/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Estudios de Casos y Controles , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Colitis/inducido químicamente , Colon/metabolismo , Ciclooxigenasa 2 , Sulfato de Dextran/toxicidad , Retroalimentación , Técnica del Anticuerpo Fluorescente , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Immunoblotting , Inmunohistoquímica , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B , Fosfoproteínas/genética , Proteínas Proto-Oncogénicas c-akt , ARN Mensajero/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E , Regeneración/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Regulación hacia Arriba , Proteínas Señalizadoras YAPRESUMEN
The aim of this study was to characterize certain prostaglandin family members in the bovine corpus luteum (CL) during the estrous cycle and pregnancy. The CL tissue was assigned to the stages 1-2, 3-4, 5-7, 8-12, 13-16 and >18 days (after regression) of the estrous cycle and 1-2, 3-4, 6-7, and >8 months of pregnancy. In these samples, we investigated prostaglandin F2alpha (PTGF), prostaglandin E2 (PTGE), their receptors (PTGFR, PTGER2, and PTGER4), cyclooxygenase 2 (COX-2), PTGF synthase (PTGFS), and PTGE synthase (PTGES). The expression of messenger RNA (mRNA) was measured by reverse transcription quantitative polymerase chain reaction, hormones by enzyme immunoassay, and localization by immunohistochemistry. The mRNA expression of COX-2, PTGFS, and PTGES in CL during the early-luteal phase was high followed by a continuous and significant downregulation afterward, as well as during all phases of pregnancy. The concentration of PTGF in CL tissue was high during the early-luteal phase, decreased significantly in the mid-luteal phase, and increased again afterward. In contrast, the concentration of PTGE increased significantly during the late-luteal phase followed by a decrease during regression. The PTGE level increased again during late pregnancy. Immunohistochemically, the large granulose-luteal cells show strong staining for COX-2 and PTGES during the early-luteal stage followed by lower activity afterward. During pregnancy, most of the luteal cells were only weakly positive or negative. In conclusion, our results indicate that the examined prostaglandin family members are involved in the local mechanisms that regulate luteal function, specifically during CL formation, function, and regression and during pregnancy in the cow.
Asunto(s)
Cuerpo Lúteo/metabolismo , Ciclooxigenasa 2/biosíntesis , Dinoprost/biosíntesis , Dinoprostona/biosíntesis , Ciclo Estral/fisiología , Hidroxiprostaglandina Deshidrogenasas/biosíntesis , Prostaglandina-E Sintasas/biosíntesis , Animales , Bovinos , Ciclooxigenasa 2/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Hidroxiprostaglandina Deshidrogenasas/genética , Fase Luteínica/metabolismo , Embarazo , Prostaglandina-E Sintasas/genética , ARN Mensajero/genética , Receptores de Prostaglandina/biosíntesisRESUMEN
Heat stress (HS) negatively affects reproduction in cattle; however, its effect on endocrine function in bovine endometrial cells remains unclear. In this study, we examined the effects of HS on the production of prostaglandin (PG) E2 and PGF2α in the cultured bovine endometrial epithelial and stromal cells separately. To evaluate the effect of HS on endocrine function, the cells were cultured at 38.5°C (control) or 40.5°C (HS). After treatment, PGE2 and PGF2α levels were measured via enzyme immunoassay (EIA) and mRNA expressions of enzymes involved in PG synthesis were examined via quantitative reverse transcription polymerase chain reaction (RT-PCR). HS did not influence the production of PGE2 or PGF2α in the epithelial cells; however, HS significantly enhanced the production of both PGE2 and PGF2α in the stromal cells (P < 0.05). In addition, HS significantly increased phospholipase A2 (PLA2), cyclooxygenase 2 (COX2), prostaglandin F synthase (PGFS), prostaglandin E synthase (PGES), and carbonyl reductase 1 (CBR1) mRNA expression in the stromal cells (P < 0.05). The overall results suggest that HS induces mRNA expression of enzymes involved in PG synthesis, resulting in the upregulation of PGE2 and PGF2α production in the stromal cells, but not in the epithelial cells. The HS-induced increase of PGE2 and PGF2α secretion in bovine endometrial stromal cells may disrupt the normal estrous cycle and cause infertility in cows during summer.