Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 351
Filtrar
1.
J Gastroenterol Hepatol ; 37(9): 1776-1784, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35877192

RESUMO

BACKGROUND AND AIM: Chronic enteropathy associated with the solute carrier organic anion transporter family member 2A1 (SLCO2A1), or CEAS, causes anemia and hypoalbuminemia in young people. Dysfunction of the SLCO2A1 transporter protein is thought to involve genetic mutation, but mutant proteins have not been functionally characterized. We examined the prostaglandin E2 (PGE2 ) transport ability of recombinant SLCO2A1 proteins containing 11 SLCO2A1 mutations found in CEAS patients. METHODS: Wild-type and mutant SLCO2A1 proteins were forcibly expressed in Xenopus laevis oocytes, and measurements of PGE2 uptake and transport capacity were compared. The membrane protein topology and functionality of the eight SLCO2A1 mutations involving single-nucleotide substitutions were predicted using computer analysis. RESULTS: The extent of functional disruption of the 11 SLCO2A1 mutations identified in CEAS patients was variable, with 10 mutations (421GT, 547GA, 664GA, 770GA, 830dupT, 830delT, 940 + 1GA, 1372GT, 1647GT, and 1807CT) resulting in loss or reduction of PGE2 transport, excluding 97GC. CONCLUSION: PGE2 transport ability of recombinant SLCO2A1 in X. laevis oocytes was hindered in 10/11 SLCO2A1 mutations identified in patients with CEAS. Further studies on the relationships between the different mutations and PGE2 transport and clinical features, such as severity, are needed.


Assuntos
Doenças Inflamatórias Intestinais , Transportadores de Ânions Orgânicos , Dinoprostona/genética , Dinoprostona/metabolismo , Humanos , Mutação , Transportadores de Ânions Orgânicos/genética
2.
J Clin Invest ; 132(9)2022 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-35499079

RESUMO

Obesity-associated complications are causing increasing morbidity and mortality worldwide. Expansion of adipose tissue in obesity leads to a state of low-grade chronic inflammation and dysregulated metabolism, resulting in insulin resistance and metabolic syndrome. Adipose tissue macrophages (ATMs) accumulate in obesity and are a source of proinflammatory cytokines that further aggravate adipocyte dysfunction. Macrophages are rich sources of cyclooxygenase (COX), the rate limiting enzyme for prostaglandin E2 (PGE2) production. When mice were fed a high-fat diet (HFD), ATMs increased expression of COX-2. Selective myeloid cell COX-2 deletion resulted in increased monocyte recruitment and proliferation of ATMs, leading to increased proinflammatory ATMs with decreased phagocytic ability. There were increased weight gain and adiposity, decreased peripheral insulin sensitivity and glucose utilization, increased adipose tissue inflammation and fibrosis, and abnormal adipose tissue angiogenesis. HFD pair-feeding led to similar increases in body weight, but mice with selective myeloid cell COX-2 still exhibited decreased peripheral insulin sensitivity and glucose utilization. Selective myeloid deletion of the macrophage PGE2 receptor subtype, EP4, produced a similar phenotype, and a selective EP4 agonist ameliorated the metabolic abnormalities seen with ATM COX-2 deletion. Therefore, these studies demonstrated that an ATM COX-2/PGE2/EP4 axis plays an important role in inhibiting adipose tissue dysfunction.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Resistência à Insulina , Tecido Adiposo/metabolismo , Animais , Ciclo-Oxigenase 2/genética , Dinoprostona/genética , Dinoprostona/metabolismo , Glucose/metabolismo , Inflamação/metabolismo , Resistência à Insulina/fisiologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/metabolismo
3.
Oncol Rep ; 47(6)2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35543149

RESUMO

Inflammatory signaling through prostaglandin E2 receptor subtype 2 (EP2) is associated with malignant tumor growth in both experimental models and cancer patients. Thus, the absence of EP2 receptors in host tissues appears to reduce tumor growth and systemic inflammation by inducing major alterations in gene expression levels across tumor tissue compartments. However, it is not yet well­established how signaling pathways in tumor tissue relate to simultaneous signaling alterations in the surrounding tumor­stroma, at conditions of reduced disease progression due to decreased host inflammation. In the present study, wild­type tumor cells, producing high levels of prostaglandin E2 (MCG 101 cells, EP2+/+), were inoculated into EP2 knockout (EP2­/­) and EP2 wild­type (EP2+/+) mice. Solid tumors were dissected into tumor­ and tumor­stroma tissue compartments for RNA expression microarray screening, followed by metabolic pathway analyses. Immunohistochemistry was used to confirm adequate dissections of tissue compartments, and to assess cell proliferation (Ki­67), prostaglandin enzymes (cyclooxygenase 2) and immunity biomarkers (CD4 and CD8) at the protein level. Microarray analyses revealed statistically significant alterations in gene expression in the tumor­stroma compartment, while significantly less pathway alterations occurred in the tumor tissue compartment. The host knockout of EP2 receptors led to a significant downregulation of cell cycle regulatory factors in the tumor­stroma compartment, while interferon γ­related pathways, chemokine signaling pathways and anti­tumor chemokines [chemokine (C­X­C motif) ligand 9 and 10] were upregulated in the tumor compartment. Thus, such gene alterations were likely related to reduced tumor growth in EP2­deficient hosts. On the whole, pathway analyses of both tumor­ and tumor­stroma compartments suggested that absence of host EP2 receptor signaling reduces 'remodeling' of tumor microenvironments and increase local immunity, probably by decreased productions of stimulating growth factors, perhaps similar to well­recognized physiological observations in wound healing.


Assuntos
Neoplasias , Receptores de Prostaglandina E Subtipo EP2 , Animais , Dinoprostona/genética , Dinoprostona/metabolismo , Humanos , Inflamação/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias/genética , Receptores de Prostaglandina E Subtipo EP2/genética , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Transdução de Sinais , Microambiente Tumoral/genética
4.
J Biol Chem ; 298(3): 101603, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35101442

RESUMO

Toll-like receptors (TLRs) are pattern recognition receptors that play a critical role in innate immune diseases. TLR3, which is localized in the endosomal compartments of hematopoietic immune cells, is able to recognize double-stranded RNA (dsRNA) derived from viruses and bacteria and thereby induce innate immune responses. Inflammatory periodontal bone resorption is caused by bacterial infections, which initially is regulated by innate immunity; however, the roles of TLR3 signaling in bone resorption are still not known. We examined the roles of TLR3 signaling in bone resorption using poly(I:C), a synthetic dsRNA analog. In cocultures of mouse bone marrow cells and stromal osteoblasts, poly(I:C) clearly induced osteoclast differentiation. In osteoblasts, poly(I:C) increased PGE2 production and upregulated the mRNA expression of PGE2-related genes, Ptgs2 and Ptges, as well as that of a gene related to osteoclast differentiation, Tnfsf11. In addition, we found that indomethacin (a COX-2 inhibitor) or an antagonist of the PGE2 receptor EP4 attenuated the poly(I:C)-induced PGE2 production and subsequent Tnfsf11 expression. Poly(I:C) also prolonged the survival of the mature osteoclasts associated with the increased mRNA expression of osteoclast marker genes, Nfatc1 and Ctsk. In ex vivo organ cultures of periodontal alveolar bone, poly(I:C) induced bone-resorbing activity in a dose-dependent manner, which was attenuated by the simultaneous administration of either indomethacin or an EP4 antagonist. These data suggest that TLR3 signaling in osteoblasts controls PGE2 production and induces the subsequent differentiation and survival of mature osteoclasts. Endogenous TLR3 in stromal osteoblasts and osteoclasts synergistically induces inflammatory alveolar bone resorption in periodontitis.


Assuntos
Reabsorção Óssea , Dinoprostona , Osteoblastos , Receptor 3 Toll-Like , Animais , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Células Cultivadas , Dinoprostona/biossíntese , Dinoprostona/genética , Dinoprostona/metabolismo , Endossomos/metabolismo , Indometacina/farmacologia , Camundongos , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Prostaglandinas E/efeitos adversos , Prostaglandinas E/metabolismo , Ligante RANK/metabolismo , RNA Mensageiro/metabolismo , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/metabolismo
5.
Bosn J Basic Med Sci ; 22(1): 100-109, 2022 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-34998360

RESUMO

Cystitis glandularis is characterized by chronic inflammation and hyperproliferation of bladder mucosa, and contributes to progression of bladder adenocarcinoma. TPRG1 (Tumor Protein P63 Regulated 1) is related to cellular inflammatory response, and dysregulation of TPRG1 in tumor tissues is associated with tumor early recurrence. The effect of TPRG1 on cystitis glandularis was investigated in this study. Firstly, bladder specimen were isolated from patients with cystitis glandularis and E. coli-induced cystitis rat. Expression of TPRG1 was found to be up-regulated in the bladder specimen. Moreover, adeno-associated virus (AAV)-mediated silence of TPRG1 was delivered into rat, and data from hematoxylin and eosin (H and E) staining showed that injection with AAV-shTPRG1 ameliorated E. coli-induced histological changes in bladder tissues of rats, and suppressed the inflammatory response. Secondly, TPRG1 was also increased in primary cystitis glandularis cells. Knockdown of TPRG1 decreased cell proliferation of primary cystitis glandularis cells, and suppressed the migration. Thirdly, cyclooxygenase-2 (COX-2) was up-regulated in the bladder specimen isolated from patients with cystitis glandularis and E. coli-induced cystitis rat. Injection with AAV-shTPRG1 reduced protein expression of COX-2, p65 and prostaglandin E2 (PGE2) in the bladder specimen. Lastly, interference of COX-2 attenuated TPRG1 over-expression-induced increase of cell proliferation and migration in the primary cystitis glandularis cells. In conclusion, TPRG1 promoted inflammation and cell proliferation of cystitis glandularis through activation of NF-кB/COX2/PGE2 axis.


Assuntos
Cistite , Proteínas/genética , Neoplasias da Bexiga Urinária , Animais , Proliferação de Células , Ciclo-Oxigenase 2/genética , Cistite/genética , Cistite/patologia , Dinoprostona/genética , Escherichia coli , Humanos , Inflamação , NF-kappa B/genética , Recidiva Local de Neoplasia , Ratos , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/patologia
6.
J Ethnopharmacol ; 282: 114645, 2022 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-34530094

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Essential oil (EO) is the main extract of patchouli and tangerine peel with antiinflammatory, antiulcer, and other functions. However, the efficacy and mechanism of the combination of EO from patchouli and tangerine peel against gastric ulcer (GU) are unclear. AIM OF THE STUDY: This study aims to reveal the protective effect of the combination of EO from patchouli and tangerine peel against GU in rats, as well as explore the optimal ratio and possible mechanism of EO in GU treatment. MATERIALS AND METHODS: The GU model is executed via water immersion and restraint stress. The repair effect of EO in different proportions on gastric mucosa injury and the effects on serum gastrin (GAS), pepsinogen C (PGC), prostaglandin E2 (PGE2), and 5-hydroxytryptamine in GU rats were observed. The optimal ratio obtained was used in the second part to set different dose groups for further experiment. The effects of the different EO doses on gastric mucosal ulcer formation and gastric acid secretion were evaluated. The morphology of chief and parietal cells were observed via transmission electron microscopy. The contents of GAS, PGC, substance P (SP), cyclic adenosine monophosphate (cAMP), cyclic guanosine monophosphate (cGMP), cholecystokinin (CCK), PGE2, and motilin (MTL) in serum in different groups were detected via enzyme-linked immunosorbent assay. Expressions of epidermal growth factor (EGF) and trefoil factor 2 (TFF2) protein in gastric tissues were detected via immunohistochemistry, and expressions of c-Jun N-terminal kinase (JNK), P53, Bcl-2-associated X protein (Bax), and Caspase-3 protein in gastric tissues were detected via western blotting. RESULTS: The EO from patchouli and tangerine peel at 1:2 ratio of compatibility significantly improved gastric mucosal injury, decreased serum GAS and PGC contents, and increased the PGE2 level in serum (p < 0.05). The mixture of EO from patchouli and tangerine peel (Mix-EO) can reduce the formation of gastric mucosal ulcers, reduce gastric mucosal injury, improve the expansion of the endoplasmic reticulum of the chief cells, repair mitochondrial damage, and inhibit the secretion of gastric acid by parietal cells. Mix-EO at 300 mg/kg can reduce the expression of serum GAS, PGC, SP, CCK, and cAMP/cGMP (p < 0.05 or 0.01); increase the expression of EGF and TFF2 protein in gastric tissues (p < 0.01); and inhibit the expression of JNK, p53, Bax, and Caspase-3 proteins (p < 0.01). CONCLUSION: The combination of EO from patchouli and tangerine peel can repair the gastric mucosal damage in GU rats and prevent the occurrence of ulcers by inhibiting the secretion of gastric acid, enhancing the defensive ability of gastric mucosa, and suppressing the apoptosis of gastric epithelial cells. Moreover, the optimal compatible ratio of patchouli and tangerine peel is 1:2.


Assuntos
Citrus/química , Óleos de Plantas/farmacologia , Pogostemon/química , Úlcera Gástrica/tratamento farmacológico , Animais , Dinoprostona/sangue , Dinoprostona/genética , Dinoprostona/metabolismo , Gastrinas/sangue , Gastrinas/genética , Gastrinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Pepsinogênio C/sangue , Pepsinogênio C/genética , Pepsinogênio C/metabolismo , Óleos de Plantas/química , Substâncias Protetoras/farmacologia , Ratos , Ratos Sprague-Dawley , Restrição Física/efeitos adversos , Serotonina/sangue , Serotonina/genética , Serotonina/metabolismo , Úlcera Gástrica/etiologia
7.
PLoS One ; 16(8): e0254606, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34428217

RESUMO

Acetylsalicylic acid is a globally used non-steroidal anti-inflammatory drug (NSAID) with diverse pharmacological properties, although its mechanism of immune regulation during inflammation (especially at in vivo relevant doses) remains largely speculative. Given the increase in clinical perspective of Acetylsalicylic acid in various diseases and cancer prevention, this study aimed to investigate the immunomodulatory role of physiological Acetylsalicylic acid concentrations (0.005, 0.02 and 0.2 mg/ml) in a human whole blood of infection-induced inflammation. We describe a simple, highly reliable whole blood assay using an array of toll-like receptor (TLR) ligands 1-9 in order to systematically explore the immunomodulatory activity of Acetylsalicylic acid plasma concentrations in physiologically relevant conditions. Release of inflammatory cytokines and production of prostaglandin E2 (PGE2) were determined directly in plasma supernatant. Experiments demonstrate for the first time that plasma concentrations of Acetylsalicylic acid significantly increased TLR ligand-triggered IL-1ß, IL-10, and IL-6 production in a dose-dependent manner. In contrast, indomethacin did not exhibit this capacity, whereas cyclooxygenase (COX)-2 selective NSAID, celecoxib, induced a similar pattern like Acetylsalicylic acid, suggesting a possible relevance of COX-2. Accordingly, we found that exogenous addition of COX downstream product, PGE2, attenuates the TLR ligand-mediated cytokine secretion by augmenting production of anti-inflammatory cytokines and inhibiting release of pro-inflammatory cytokines. Low PGE2 levels were at least involved in the enhanced IL-1ß production by Acetylsalicylic acid.


Assuntos
Aspirina/farmacologia , Citocinas/genética , Inflamação/tratamento farmacológico , Receptores Toll-Like/genética , Adjuvantes Imunológicos/farmacologia , Adolescente , Adulto , Anti-Inflamatórios não Esteroides/farmacologia , Celecoxib/farmacologia , Ciclo-Oxigenase 2/sangue , Ciclo-Oxigenase 2/genética , Citocinas/biossíntese , Dinoprostona/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Indometacina/farmacologia , Inflamação/sangue , Inflamação/patologia , Interleucina-10/genética , Interleucina-1beta/genética , Interleucina-6/genética , Leucócitos/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Receptores Toll-Like/sangue , Adulto Jovem
8.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1866(12): 159032, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34428549

RESUMO

N-3 polyunsaturated fatty acids (n-3 PUFAs), and in particular docosahexaenoic acid (DHA), have many beneficial metabolic effects, including reducing epidermal thickness in patients with psoriasis. The positive impacts of DHA in psoriasis could be mediated by its interactions with the PPAR signaling pathway, as well as by its secretion of anti-inflammatory bioactive metabolites, but the detailed metabolism is still not understood. In the present study, we evaluated the influence of DHA on the main features of psoriasis and its effects on the PPAR signaling pathway, in a psoriatic in vitro skin model. Healthy and psoriatic skin substitutes were produced according to the tissue-engineered self-assembly method, using culture media supplemented with 10 µM of DHA. The presence of DHA led to a reduction in the abnormal cell differentiation of psoriatic keratinocytes, seen in the increased expression of filaggrin and keratin 10. DHA was incorporated into the membrane phospholipids of the epidermis and transformed principally into eicosapentaenoic acid (EPA). Furthermore, the addition of DHA into the culture medium led to a decrease in the levels of lipid mediators derived from n-6 PUFAs, mainly prostaglandin E2 (PGE2) and 12-hydroxyeicosatetraenoic acid (12-HETE). Finally, DHA supplementation rebalanced the expression of PPAR receptors and caused a decrease in the secretion of TNF-α. Altogether, our results show that DHA possesses the ability to attenuate the psoriatic characteristics of psoriatic skin substitutes, mostly by restoring epidermal cell differentiation and proliferation, as well as by reducing inflammation.


Assuntos
Ácidos Docosa-Hexaenoicos/metabolismo , Ácidos Graxos Ômega-3/metabolismo , Psoríase/metabolismo , Pele/metabolismo , Adolescente , Adulto , Biópsia , Técnicas de Cultura de Células , Diferenciação Celular/genética , Dinoprostona/genética , Ácido Eicosapentaenoico/análogos & derivados , Ácido Eicosapentaenoico/farmacologia , Feminino , Proteínas Filagrinas/genética , Humanos , Queratina-10/genética , Queratinócitos/metabolismo , Queratinócitos/patologia , Metabolismo dos Lipídeos/genética , Masculino , Pessoa de Meia-Idade , PPAR alfa/genética , PPAR gama/genética , Psoríase/genética , Psoríase/patologia , Pele/patologia , Adulto Jovem
9.
Cancer Lett ; 518: 180-195, 2021 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-34216690

RESUMO

Human anterior gradient homolog 2 (AGR2) reportedly acts as an oncogene in multiple types of cancers. As a secreted protein, the oncogenic roles of extracellular AGR2 have been the focus of the increasing number of studies. In contrast, the oncological functions of intracellular AGR2 (iAGR2) remain elusive. Here, we report that intracellular AGR2 (iAGR2) is sufficient to promote CRC metastasis. iAGR2 binds to KDEL receptors (KDELRs) via its KTEL motif to activate downstream Gs-PKA signaling. Activated PKA upregulates the expression of NF-κB subunit c-Rel (REL) and acetylates histone H3 at lysine 9 (H3K9ac) to promote the transcription of SNAIL and SLUG. AGR2 can be upregulated by prostaglandin E2 (PGE2) via EP4-PI3K-AKT pathway and is indispensable for PGE2-induced CRC metastasis. AGR2 knockdown enhances therapeutic effects of a COX-2 inhibitor, celecoxib, in CRC metastasis. Collectively, our study reveals a promoting role and molecular mechanisms of iAGR2 in CRC metastasis and uncovers a new tumor microenvironment signal regulating AGR2 expression, which may provide new targets for treating metastatic CRC.


Assuntos
Neoplasias Colorretais/genética , Dinoprostona/genética , Transição Epitelial-Mesenquimal/genética , Mucoproteínas/genética , Proteínas Oncogênicas/genética , Animais , Celecoxib/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Células HCT116 , Células HEK293 , Células HT29 , Histonas/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Oncogenes/genética , Fosfatidilinositol 3-Quinases/genética , Receptores de Peptídeos/genética , Transdução de Sinais/genética , Microambiente Tumoral/genética
10.
Sci Rep ; 11(1): 10770, 2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-34031466

RESUMO

In periodontitis, gingival fibroblasts (GFs) interact with and respond to oral pathogens, significantly contributing to perpetuation of chronic inflammation and tissue destruction. The aim of this study was to determine the usefulness of the recently released hTERT-immortalized GF (TIGF) cell line for studies of host-pathogen interactions. We show that TIGFs are unable to upregulate expression and production of interleukin (IL)-6, IL-8 and prostaglandin E2 upon infection with Porphyromonas gingivalis despite being susceptible to adhesion and invasion by this oral pathogen. In contrast, induction of inflammatory mediators in TNFα- or IL-1ß-stimulated TIGFs is comparable to that observed in primary GFs. The inability of TIGFs to respond directly to P. gingivalis is caused by a specific defect in Toll-like receptor-2 (TLR2) expression, which is likely driven by TLR2 promoter hypermethylation. Consistently, TIGFs fail to upregulate inflammatory genes in response to the TLR2 agonists Pam2CSK4 and Pam3CSK4. These results identify important limitations of using TIGFs to study GF interaction with oral pathogens, though these cells may be useful for studies of TLR2-independent processes. Our observations also emphasize the importance of direct comparisons between immortalized and primary cells prior to using cell lines as models in studies of any biological processes.


Assuntos
Infecções por Bacteroidaceae/imunologia , Gengiva/citologia , Interleucina-1beta/genética , Porphyromonas gingivalis/patogenicidade , Telomerase/genética , Fator de Necrose Tumoral alfa/genética , Aderência Bacteriana/efeitos dos fármacos , Infecções por Bacteroidaceae/genética , Células Cultivadas , Metilação de DNA , Dinoprostona/genética , Dinoprostona/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/imunologia , Fibroblastos/metabolismo , Gengiva/efeitos dos fármacos , Gengiva/imunologia , Gengiva/metabolismo , Humanos , Interleucina-1beta/metabolismo , Lipopeptídeos/farmacologia , Oligopeptídeos/farmacologia , Receptor 2 Toll-Like/agonistas , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Receptor Toll-Like 9/agonistas , Fator de Necrose Tumoral alfa/metabolismo
11.
Eur J Pharmacol ; 906: 174173, 2021 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-34033814

RESUMO

According to numerous epidemiological studies, aspirin is a non-steroidal anti-inflammatory drug (NSAID) that reduces the occurrence and mortality of colorectal cancer (CRC). However, the underlying mechanisms are not well identified. In an effort to fill these gaps, we administered aspirin on mice one day before induction in an azoxymethane (AOM)/dextran sulfate sodium (DSS) induced CRC model. In this study, we assessed the effects of aspirin on tumorigenesis and tumor cell proliferation. Multi-layer analyses were carried out to identify changes in cytokines, metabolites, level of gene expressions, and proteins associated with tumorigenesis and aspirin treatment. The results showed that aspirin-treated mice developed fewer colon tumors in response to AOM/DSS, and aspirin can actively block cyclooxygenase (COX) metabolism and reduce levels of pro-inflammatory cytokines. In addition, the transcriptomic and proteomic analyses both indicated that aspirin has an inhibitory effect on the Wnt pathway. The in vitro results further indicated that aspirin inhibits WNT6 production, possibly by suppressing its transcription factor NR4A2, which in turn is regulated by prostaglandin E2, thereby ultimately inhibiting the Wnt pathway. These findings improve our understanding of the mechanisms behind aspirin's chemoprevention effect on CRC.


Assuntos
Aspirina/farmacologia , Neoplasias Associadas a Colite/prevenção & controle , Colite/tratamento farmacológico , Dinoprostona/antagonistas & inibidores , Animais , Aspirina/uso terapêutico , Azoximetano/administração & dosagem , Azoximetano/toxicidade , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Colite/induzido quimicamente , Colite/patologia , Neoplasias Associadas a Colite/patologia , Sulfato de Dextrana/administração & dosagem , Sulfato de Dextrana/toxicidade , Dinoprostona/genética , Dinoprostona/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Humanos , Masculino , Camundongos , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/antagonistas & inibidores , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Proteômica , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos
12.
PLoS One ; 16(5): e0251942, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34003864

RESUMO

Prostaglandin E2 receptor EP4 is involved in inflammation and related tumorigenesis in the colorectum. This study aimed to investigate the chemopreventive ability of RQ-15986, a selective EP4 antagonist, in colitis-related colorectal tumorigenesis. Male Kyoto APC delta rats, which have APC mutations, were treated with azoxymethane and dextran sulfate sodium and subsequently administered RQ-15986 for eight weeks. At the end of the experiment, the development of colorectal tumor was significantly inhibited in the RQ-15986-treated group. The cell proliferation of the crypts and tumors in the colorectum was decreased following RQ-15986 treatment. RQ-15986 also suppressed the expression of pro-inflammatory cytokines, including tumor necrosis factor-α, interleukin-6, interleukin-18, and monocyte chemotactic protein-1, in the colon mucosa. In addition, the expression levels of indoleamine 2,3-dioxygenase, which is involved in immune tolerance, were decreased in the colorectal epithelium and tumors of the RQ-15986-treated group. These findings indicate that RQ-15986 inhibits colitis-associated colorectal tumorigenesis by attenuating inflammation, suppressing cell proliferation, and modulating the expression of indoleamine 2,3-dioxygenase. Targeting prostaglandin E2/EP4 signaling might be a useful strategy for chemoprevention of inflammation-related colorectal cancer.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Neoplasias do Colo/tratamento farmacológico , Dinoprostona/genética , Inflamação/tratamento farmacológico , Receptores de Prostaglandina E Subtipo EP4/genética , Animais , Azoximetano/toxicidade , Benzamidas/farmacologia , Carcinogênese/genética , Proliferação de Células/efeitos dos fármacos , Quimiocina CCL2/genética , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Sulfato de Dextrana/toxicidade , Dinoprostona/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/patologia , Interleucina-18/genética , Interleucina-6/genética , Mutação/genética , Ratos , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética
13.
Med Mycol ; 59(9): 869-881, 2021 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-33862618

RESUMO

Components of the iron reductive pathway of Candida albicans have been implicated in the production of prostaglandin E2 (PGE2) and virulence. However, it is unknown whether other components of this pathway influence PGE2. We investigated the role of the iron reductive pathway of C. albicans in biofilm formation, PGE2 production, and virulence in Caenorhabditis elegans. Additionally, as the co-occurrence of C. albicans and Pseudomonas aeruginosa in host tissues is frequent and involves competition for host-associated iron, we examined the effects of this interaction. Deletion of multicopper oxidase gene, FET99, and iron permease genes, FTH1 and FTH2, affected biofilm metabolic activity, and for the FTH2 mutant, also biofilm morphology. Deletion of CCC1 (vacuolar iron transporter) and CCC2 (P-type ATPase copper importer) also influenced biofilm morphology. For PGE2 production, deletion of FET99, FTH1, FTH2, CCC1, and CCC2 caused a significant reduction by monomicrobial biofilms, while FTH2deletion caused the highest reduction in polymicrobial biofilms. URA3 positive mutants of FET99 and FTH2 demonstrated attenuated virulence in C. elegans, potentially due to the inability of mutants to form hyphae in vivo. Deductively, the role of the iron reductive pathway in PGE2 synthesis is indirect, possibly due to their role in iron homeostasis. LAY SUMMARY: Iron uptake is vital for disease-causing microbes like Candida albicans. Using strains deficient in some iron-uptake genes, we show that iron-uptake genes, especially FET99 and FTH2, play a role in biofilm formation, prostaglandin production, and virulence in the nematode infection model.


Assuntos
Biofilmes/efeitos dos fármacos , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/parasitologia , Candida albicans/metabolismo , Dinoprostona/biossíntese , Ferro/metabolismo , Pseudomonas aeruginosa/metabolismo , Virulência/efeitos dos fármacos , Animais , Candida albicans/genética , Candida albicans/patogenicidade , Dinoprostona/genética , Variação Genética , Genótipo , Interações Hospedeiro-Patógeno/genética , Redes e Vias Metabólicas/genética , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidade , Virulência/genética
14.
Pol J Vet Sci ; 24(1): 5-12, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33847092

RESUMO

Dieldrin and DDE are environmental metabolites of the organochlorine pesticides aldrin and DDT, respectively. During pregnancy, these chemicals can quickly infiltrate through the placental barrier, accumulate in amniotic fluid and fetus, and act as endocrine disruptors (EDs). The aim of this study was to investigate the effect of DDE and dieldrin and their parental substances at concentrations of 1 and 10 ng/ml on secretion of PGE2 and PGF2α from bovine endometrial explants (120-150 and 151-180 days of pregnancy) after 24 hr of incubation with EDs. The mRNA expression of COX2, PGES and PGFS and the concentrations of PGE2 and PGF2α were measured. EDs did not affect (p>0.05) COX2 gene expression, but DDT and DDE decreased (p⟨0.05) PGES expression and PGE2 secretion in the explants from 120-150 days of pregnancy. Depending on the dose, DDT and DDE increased (p⟨0.05) PGFS expression and PGF2α secretion from the explants from 120-150 days and decreased PGF2α secretion (p⟨0.05) from the explants from 151-180 days of pregnancy. Aldrin and dieldrin decreased (p⟨0.05) PGFS expression and PGF2α secretion from all explants. In summary, EDs disrupt the secretion of PGE2 and PGF2α by influencing the gene expression of PGES and PGFS.


Assuntos
Bovinos/fisiologia , Dinoprosta/metabolismo , Dinoprostona/metabolismo , Endométrio/efeitos dos fármacos , Inseticidas/farmacologia , Aldrina/farmacologia , Aldrina/toxicidade , Animais , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , DDT/farmacologia , DDT/toxicidade , Diclorodifenil Dicloroetileno/farmacologia , Diclorodifenil Dicloroetileno/toxicidade , Dieldrin/farmacologia , Dieldrin/toxicidade , Dinoprosta/genética , Dinoprostona/genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Inseticidas/metabolismo , Inseticidas/toxicidade , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Técnicas de Cultura de Tecidos/veterinária
15.
Radiat Res ; 195(2): 115-127, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33302300

RESUMO

Identification of medical countermeasures (MCM) to mitigate radiation damage and/or protect first responders is a compelling unmet medical need. The prostaglandin E2 (PGE2) analog, 16,16 dimethyl-PGE2 (dmPGE2), has shown efficacy as a radioprotectant and radiomitigator that can enhance hematopoiesis and ameliorate intestinal mucosal cell damage. In this study, we optimized the time of administration of dmPGE2 for protection and mitigation against mortality from the hematopoietic acute radiation syndrome (H-ARS) in young adult mice, evaluated its activity in pediatric and geriatric populations, and investigated potential mechanisms of action. Windows of 30-day survival efficacy for single administration of dmPGE2 were defined as within 3 h prior to and 6-30 h after total-body γ irradiation (TBI). Radioprotective and radio-mitigating efficacy was also observed in 2-year-old geriatric mice and 6-week-old pediatric mice. PGE2 receptor agonist studies suggest that signaling through EP4 is primarily responsible for the radioprotective effects. DmPGE2 administration prior to TBI attenuated the drop in red blood cells and platelets, accelerated recovery of all peripheral blood cell types, and resulted in higher hematopoietic and mesenchymal stem cells in survivor bone marrow. Multiplex analysis of bone marrow cytokines together with RNA sequencing of hematopoietic stem cells indicated a pro-hematopoiesis cytokine milieu induced by dmPGE2, with IL-6 and G-CSF strongly implicated in dmPGE2-mediated radioprotective activity. In summary, we have identified windows of administration for significant radio-mitigation and radioprotection by dmPGE2 in H-ARS, demonstrated survival efficacy in special populations, and gained insight into radioprotective mechanisms, information useful towards development of dmPGE2 as a MCM for first responders, military personnel, and civilians facing radiation threats.


Assuntos
Síndrome Aguda da Radiação/tratamento farmacológico , Dinoprostona/farmacologia , Tolerância a Radiação/genética , Protetores contra Radiação/farmacologia , Síndrome Aguda da Radiação/genética , Síndrome Aguda da Radiação/patologia , Animais , Dinoprostona/análogos & derivados , Dinoprostona/genética , Relação Dose-Resposta à Radiação , Raios gama/efeitos adversos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Fator Estimulador de Colônias de Granulócitos/genética , Hematopoese/efeitos dos fármacos , Hematopoese/efeitos da radiação , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos da radiação , Humanos , Interleucina-6/genética , Camundongos , Tolerância a Radiação/efeitos dos fármacos , Análise de Sequência de RNA , Irradiação Corporal Total
16.
Z Naturforsch C J Biosci ; 76(3-4): 103-110, 2021 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-32986614

RESUMO

Chikusetsusaponin IVa (CHS-IVa), a saponin from herb Panacis japonicas, possesses extensive biological activities. However, the roles and underlying mechanisms of CHS-IVa on inflammation have not been fully clarified in the setting of murine macrophages. In this study, we found that CHS-IVa effectively reduced the expression of inflammatory mediators, including interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), interleukin-10 (IL-10), interleukin-1ß (IL-1ß), cyclooxygenase (COX-2), inducible nitric oxide synthase (iNOS) in lipopolysaccharide (LPS)-stimulated murine macrophage-like RAW264.7 cells. Meanwhile, CHS-IVa could also evidently bate the contents of nitric oxide (NO) and prostaglandin E2 (PGE2) in cell culture supernatants. Furthermore, the anti-inflammatory activity of CHS-IVa may be via diminishing the phosphorylation of extracellular regulated protein kinases (ERK), p38, and c-Jun N-terminal kinase (JNK). Collectively, these findings will help to understand of the anti-inflammatory effects and mechanisms of P. japonicas deeply, and suggest a validated therapeutic use as an anti-inflammatory medication.


Assuntos
Inflamação/tratamento farmacológico , Macrófagos/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Ácido Oleanólico/análogos & derivados , Saponinas/farmacologia , Animais , Dinoprostona/genética , Humanos , Inflamação/genética , Inflamação/patologia , Interleucina-10/genética , Interleucina-1beta/genética , Interleucina-6/genética , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Camundongos , Óxido Nítrico/genética , Ácido Oleanólico/farmacologia , Células RAW 264.7 , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética
17.
Steroids ; 165: 108759, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33181144

RESUMO

Gastric ulcers are a very common public health problem affecting up to 10% worldwide. Russelioside B is a steroidal glycoside isolated from several Caralluma species. No study tested the ulcer healing potential of the compound. The current study aimed to assess the protective effect of russelioside B against ethanol-induced gastric mucosal injury in rats. Ulcer was induced on rats by a single intragastric dose of absolute ethanol (5 mL/kg). Rats were randomly assorted into four groups (n = 8) and given treatments (Antodine, 20 mg/kg or russelioside B, 50 mg/kg) by oral gavage 1 h before ulcer induction. Pretreatment with russelioside B (50 mg/kg) attenuated the gastric mucosal injury as proved by a decrease of ulcer index, and histological scores. It suppressed the gastric inflammation by a significant lowering the tumor necrosis factor-α and interleukin-6 levels with myeloperoxidase activity (which are also aggravating factors in the case of Covid-19 infection). In addition, administration of russelioside B halted the gastric oxidative stress via inhibition of lipid peroxides by maintaining reduced glutathione and by decreasing malondialdehyde. It was able also to restore the sharp drop in the levels of heat shock protein-70, vascular endothelial growth factor and prostaglandin E2 induced by ethanol. Additionally, it showed carbonic anhydrase inhibition activity. The gastroprotective action of russelioside B was umpired through multi mechanistic actions; suppression of gastric oxidative stress, inflammation, anti-apoptotic activities and enhanced gastric mucosal protection by up-regulation of endothelial growth factor, normalization of heat shock protein-70 and prostaglandin E2. These actions were comparable in part to some classical antiulcer drugs such as Antodine.


Assuntos
Dinoprostona/genética , Glicosídeos/farmacologia , Proteínas de Choque Térmico HSP70/genética , Pregnanos/farmacologia , Úlcera Gástrica/tratamento farmacológico , Animais , Antiulcerosos/farmacologia , Apocynaceae/química , COVID-19/genética , COVID-19/virologia , Modelos Animais de Doenças , Etanol/toxicidade , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glicosídeos/química , Humanos , Interleucina-6/genética , Estresse Oxidativo/efeitos dos fármacos , Peroxidase/genética , Pregnanos/química , Ratos , SARS-CoV-2/genética , SARS-CoV-2/patogenicidade , Úlcera Gástrica/induzido quimicamente , Úlcera Gástrica/genética , Úlcera Gástrica/patologia , Fator de Necrose Tumoral alfa/genética , Tratamento Farmacológico da COVID-19
18.
Int J Mol Sci ; 21(20)2020 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-33081378

RESUMO

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.


Assuntos
Adenocarcinoma/metabolismo , Dinoprostona/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Gástricas/metabolismo , Adenocarcinoma/genética , Idoso , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/genética , Feminino , Mucosa Gástrica/metabolismo , Humanos , Hidroxiprostaglandina Desidrogenases/genética , Hidroxiprostaglandina Desidrogenases/metabolismo , Masculino , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/metabolismo , Neoplasias Gástricas/genética
19.
Anim Reprod Sci ; 221: 106549, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32861111

RESUMO

Cows nearing parturition have a negative energy balance (NEB), which is closely associated with lesser fertility. The NEB results in greater fat mobilisation and production of a large amount of non-esterified fatty acid (NEFA). Prostaglandins (PG), especially prostaglandin E2 (PGE2) and prostaglandin F2α (PGF2α), have important functions in regulating reproductive function. There, however, is little known about how the synthesis and release of PG are affected by NEFA. In this study, there was a focus on effects of NEFA on PG secretion as well as relative abundances of mRNA transcript and protein for PG synthetases and PG receptors in bovine endometrial (BEND) cells. Proliferation rate of BEND cells decreased in a concentration-dependent manner as NEFA increased in the media. The concentrations of PGE2 and PGF2α in NEFA treatment groups also decreased, while the ratio of PGE2/PGF2α and the relative abundances of proteins and mRNA that regulate PG synthesis and PG receptor mRNA transcripts and protein were greater as the NEFA concentration increased. Collectively, when there were large NEFA concentrations in the medium, there was a lesser release of PGE2 and PGF2α, however, there was a greater ratio of PGE2/PGF2α and relative abundances of mRNA transcripts and protein for PG synthetases and PG receptors in BEND cells, which changed the internal milieu and physiological function of the uterus with possible effects on fertility after calving. These findings provide important information that will help for further investigation of associations between NEB and fertility in dairy cows during the non-lactation to lactation-transition period.


Assuntos
Bovinos/fisiologia , Dinoprosta/metabolismo , Dinoprostona/metabolismo , Endométrio/citologia , Ácidos Graxos não Esterificados/farmacologia , RNA Mensageiro/metabolismo , Animais , Bovinos/genética , Linhagem Celular , Sobrevivência Celular , Dinoprosta/genética , Dinoprostona/genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , RNA Mensageiro/genética , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/metabolismo
20.
Int J Mol Sci ; 21(15)2020 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-32751738

RESUMO

Microglial hyperactivation and neuroinflammation are known to induce neuronal death, which is one of the main causes of neurodegenerative disorders. We previously found that Aquilariae Lignum extract attenuated both neuronal excitotoxicity and neuroinflammation in vivo and in vitro. For further analysis, we extracted the methylene chloride fraction of Aquilariae Lignum to determine the bioactive compounds. In this study, we investigated the anti-neuroinflammatory effects and underlying mechanisms of the Aquilariae Lignum fraction (ALF) using lipopolysaccharide (LPS)-stimulated BV2 microglial cells. BV2 cells were pretreated with ALF (0.5, 1, and 2.5 µg/mL) before treatment with LPS (1 µg/mL). Pretreatment with ALF significantly attenuated the LPS-induced overproductions of nitric oxide (NO), cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), and interleukin (IL)-1ß. These anti-inflammatory effects were supported by ALF-mediated modulation of the nuclear factor-kappa B (NF-κB) pathway. Furthermore, ALF exerted strong anti-inflammasome effects, as shown by IL-1ß-specific inhibitory activity, but not activity against tumor necrosis factor (TNF)-α, along with inhibition of caspase-1 activity and NACHT, LRR, and PYD domain-containing protein 3 (NLRP3)-related molecules. These results indicate the potent anti-neuroinflammatory activity of ALF and that its underlying mechanism may involve the regulation of NLRP3 inflammasome-derived neuroinflammation in microglial cells.


Assuntos
Anti-Inflamatórios/farmacologia , Inflamação/tratamento farmacológico , Cloreto de Metileno/farmacologia , Thymelaeaceae/química , Animais , Anti-Inflamatórios/química , Ciclo-Oxigenase 2/genética , Dinoprostona/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/genética , Humanos , Inflamação/metabolismo , Inflamação/patologia , Interleucina-1beta/genética , Lipopolissacarídeos/química , Lipopolissacarídeos/farmacologia , Cloreto de Metileno/química , Microglia/efeitos dos fármacos , Microglia/patologia , NF-kappa B/genética , Óxido Nítrico/genética , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA