Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Pharmacol Sci ; 154(2): 97-107, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38246733

RESUMEN

In our previous study, we reported that 2, 5-dimethyl-celecoxib (DM-C), a derivative of celecoxib, prevents cardiac remodeling in different mouse models of heart failure, including myocardial infarction (MI). The inflammatory response after MI affects the progression of cardiac remodeling, wherein the immune cells, mainly macrophages, play crucial roles. Therefore, we evaluated the effect of DM-C on macrophages in a cryoinjury-induced myocardial infarction (CMI) mouse model. We observed that DM-C attenuated the deterioration of left ventricular ejection fraction and cardiac fibrosis 14 d after CMI. Gene expression of pro-inflammatory cytokines at the infarct site was reduced by DM-C treatment. Analysis of macrophage surface antigens revealed that DM-C induced transient accumulation of macrophages at the infarct site without affecting their polarization. In vitro experiments using peritoneal monocytes/macrophages revealed that DM-C did not directly increase the phagocytic ability of the macrophages but increased their number, thereby upregulating the clearance capacity. Moreover, DM-C rapidly excluded the cells expressing necrotic cell marker from the infarct site. These results suggested that DM-C enhanced the clearance capacity of macrophages by transiently increasing their number at the infarct site, and terminated the escape from the inflammatory phase earlier, thereby suppressing excessive cardiac remodeling and ameliorating cardiac dysfunction.


Asunto(s)
Infarto del Miocardio , Pirazoles , Sulfonamidas , Remodelación Ventricular , Animales , Ratones , Celecoxib/farmacología , Celecoxib/uso terapéutico , Volumen Sistólico , Función Ventricular Izquierda , Infarto del Miocardio/tratamiento farmacológico , Macrófagos , Modelos Animales de Enfermedad
2.
Endocr J ; 67(1): 91-94, 2020 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-31534059

RESUMEN

Uterine sarcomas are rare and aggressive gynecologic tumors with poor prognosis; therefore, early diagnosis is crucial for therapy. However, it is very difficult to distinguish uterine sarcomas from leiomyomas which are common benign uterine tumors. Therefore, the development of a diagnostic method that utilizes reliable biomarkers to distinguish uterine sarcomas from leiomyomas is important so as to identify the rare tumors. The candidate factors as novel biomarkers were searched for in public databases and a pilot study was performed for confirmation. Growth differentiation factor-15 (GDF15), progranulin, and osteopontin were identified as candidate biomarkers for diagnosing uterine sarcoma. Thus, developing a rapid and easy method to measure these factors could help establish a screening system for uterine sarcomas. In this study, we developed a novel measurement system for these factors using a compact chemical luminescence immunological automatic analyzer POCubeTM. This assay system, which is based on the flow-through membrane immunoassay, completes the whole process and generates results within 15 min. Serum concentrations of these factors measured via POCubeTM correlated well with those measured using enzyme-linked immunosorbent assay (r = 0.994 for GDF15, r = 0.992 for progranulin, and r = 0.976 for osteopontin). The POCubeTM system provides rapid and easy measurement of these factors, thereby facilitating uterine sarcoma diagnosis.


Asunto(s)
Factor 15 de Diferenciación de Crecimiento/sangre , Leiomioma/sangre , Osteopontina/sangre , Progranulinas/sangre , Sarcoma/sangre , Neoplasias Uterinas/sangre , Diagnóstico Diferencial , Femenino , Humanos , Inmunoensayo , Leiomioma/diagnóstico , Proyectos Piloto , Curva ROC , Sarcoma/diagnóstico , Sensibilidad y Especificidad , Factores de Tiempo , Neoplasias Uterinas/diagnóstico
3.
Proc Natl Acad Sci U S A ; 112(13): 4086-91, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25775533

RESUMEN

Most patients suffering from cancer die of metastatic disease. Surgical removal of solid tumors is performed as an initial attempt to cure patients; however, surgery is often accompanied with trauma, which can promote early recurrence by provoking detachment of tumor cells into the blood stream or inducing systemic inflammation or both. We have previously reported that administration of atrial natriuretic peptide (ANP) during the perioperative period reduces inflammatory response and has a prophylactic effect on postoperative cardiopulmonary complications in lung cancer surgery. Here we demonstrate that cancer recurrence after curative surgery was significantly lower in ANP-treated patients than in control patients (surgery alone). ANP is known to bind specifically to NPR1 [also called guanylyl cyclase-A (GC-A) receptor]. In mouse models, we found that metastasis of GC-A-nonexpressing tumor cells (i.e., B16 mouse melanoma cells) to the lung was increased in vascular endothelium-specific GC-A knockout mice and decreased in vascular endothelium-specific GC-A transgenic mice compared with control mice. We examined the effect of ANP on tumor metastasis in mice treated with lipopolysaccharide, which mimics systemic inflammation induced by surgical stress. ANP inhibited the adhesion of cancer cells to pulmonary arterial and micro-vascular endothelial cells by suppressing the E-selectin expression that is promoted by inflammation. These results suggest that ANP prevents cancer metastasis by inhibiting the adhesion of tumor cells to inflamed endothelial cells.


Asunto(s)
Factor Natriurético Atrial/farmacología , Células Endoteliales/citología , Neoplasias/metabolismo , Animales , Adhesión Celular , Línea Celular Tumoral , Supervivencia sin Enfermedad , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inflamación , Estimación de Kaplan-Meier , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Melanoma Experimental , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia , Neoplasias/patología , Estudios Retrospectivos
4.
J Pharmacol Sci ; 133(4): 268-275, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28433565

RESUMEN

Extract of pine nodules (matsufushi) formed by bark proliferation on the surface of trees of Pinus tabulaeformis or Pinus massoniana has been used as an analgesic for joint pain, rheumatism, neuralgia, dysmenorrhea and other complaints in Chinese traditional medicine. Here we report the effects of matsufushi extract and its components on catecholamine secretion and synthesis in cultured bovine adrenal medullary cells. We found that matsufushi extract (0.0003-0.005%) and its component, SJ-2 (5-hydroxy-3-methoxy-trans-stilbene) (0.3-100 µM), but not the other three, concentration-dependently inhibited catecholamine secretion induced by acetylcholine, a physiological secretagogue. Matsufushi extract (0.0003-0.005%) and SJ-2 (0.3-100 µM) also inhibited 45Ca2+ influx induced by acetylcholine in a concentration-dependent manner, similar to its effect on catecholamine secretion. They also suppressed 14C-catecholamine synthesis and tyrosine hydroxylase activity induced by acetylcholine. In Xenopus oocytes expressing α3ß4 nicotinic acetylcholine receptors, matsufushi extract (0.00003-0.001%) and SJ-2 (1-100 µM) directly inhibited the current evoked by acetylcholine. The present findings suggest that SJ-2, as well as matsufushi extract, inhibits acetylcholine-induced catecholamine secretion and synthesis by suppression of nicotinic acetylcholine receptor-ion channels in bovine adrenal medullary cells.


Asunto(s)
Acetilcolina/farmacología , Médula Suprarrenal/citología , Médula Suprarrenal/metabolismo , Catecolaminas/biosíntesis , Catecolaminas/metabolismo , Pinus/química , Extractos Vegetales/química , Extractos Vegetales/farmacología , Estilbenos/farmacología , Acetilcolina/antagonistas & inhibidores , Animales , Calcio/metabolismo , Bovinos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Antagonistas Nicotínicos , Extractos Vegetales/aislamiento & purificación , Receptores Nicotínicos/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Xenopus
5.
J Surg Res ; 194(2): 631-637, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25483737

RESUMEN

BACKGROUND: C-type natriuretic peptide (CNP), secreted by vascular endothelial cells, belongs to a family of peptides that includes atrial and brain natriuretic peptides. CNP exhibits many vasoprotective effects against pulmonary hypertension and pulmonary fibrosis. The objective of this study was to investigate the prophylactic effects of CNP in a mouse model of lipopolysaccharide (LPS)-induced acute lung injury (ALI). MATERIALS AND METHODS: C57BL/6 mice were divided into three groups as follows: normal control mice (n = 13), LPS mice treated with vehicle (n = 12), and LPS mice treated with CNP (n = 12). Twenty-four hours after tail vein injection of LPS, histopathologic, gene expression, and bronchoalveolar lavage fluid (BALF) assessments were performed on the lungs. To examine the neutrophils in the lungs, cells positive for myeloperoxidase staining were detected by immunohistochemistry. BALF cytokine levels were analyzed by enzyme-linked immunosorbent assays. Gene expression in lung tissue was analyzed by real-time polymerase chain reaction. RESULTS: CNP significantly attenuated the elevation of leukocyte cell counts and levels of tumor necrosis factor-alpha, macrophage inflammatory protein-2, monocyte chemoattractant protein-1, interleukin-6, and keratinocyte-derived chemokine in the BALF after LPS injection. Furthermore, there were significantly fewer myeloperoxidase-positive cells in lungs treated with CNP after LPS injection. In lungs of CNP-treated mice, expression of the monocyte chemoattractant protein-1, S100A8, and E-selectin genes was significantly lower than that in vehicle-treated mice. CONCLUSIONS: CNP had a protective effect on ALI induced by LPS by reducing inflammatory cell infiltration. CNP may hold promise in therapeutic strategies for ALI after pulmonary resection surgery.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Péptido Natriurético Tipo-C/uso terapéutico , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/patología , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Calgranulina A/metabolismo , Quimiocina CCL2/metabolismo , Evaluación Preclínica de Medicamentos , Selectina E/metabolismo , Lipopolisacáridos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Péptido Natriurético Tipo-C/farmacología
6.
Endocr J ; 62(9): 757-63, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26135521

RESUMEN

Steroid hormones are synthesized from cholesterol in various tissues, mainly in the adrenal glands and gonads. Because these lipid-soluble steroid hormones immediately diffuse through the cells in which they are produced, their secretion directly reflects the activity of the genes related to their production. Progesterone is important not only for luteinization and maintenance of pregnancy, but also as a substrate for most other steroids. Steroidogenic acute regulatory protein (STAR), cytochrome P450 cholesterol side-chain cleavage enzyme (P450scc), and 3ß-hydroxysteroid dehydrogenase/Δ(5)-Δ(4) isomerase (3ß-HSD) are well-known proteins essential for progesterone production. In addition to them, glutathione S-transferase A1-1 and A3-3 are shown to exert Δ(5)-Δ(4) isomerization activity to produce progesterone in a cooperative fashion with 3ß-HSD. 5-Aminolevulinic acid synthase 1, ferredoxin 1, and ferredoxin reductase also play a role in steroidogenesis as accessory factors. Members of the nuclear receptor 5A (NR5A) family (steroidogenic factor 1 and liver receptor homolog 1) play a crucial role in the transcriptional regulation of these genes. The NR5A family activates these genes by binding to NR5A responsive elements present within their promoter regions, as well as to the elements far from their promoters. In addition, various NR5A-interacting proteins including peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), nuclear receptor subfamily 0, group B, member 1 (DAX-1), and CCAAT/enhancer-binding proteins (C/EBP) are involved in the transcription of NR5A target genes and regulate the transcription either positively or negatively under both basal and tropic hormone-stimulated conditions. In this review, we describe the transcriptional regulation of genes related to progesterone production.


Asunto(s)
Regulación de la Expresión Génica , Progesterona/biosíntesis , Transcripción Genética , 17-Hidroxiesteroide Deshidrogenasas/genética , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Glutatión Transferasa/genética , Humanos , Fosfoproteínas/genética , Regiones Promotoras Genéticas , Factor Esteroidogénico 1/genética
7.
Biochem J ; 460(3): 459-71, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24635384

RESUMEN

The transcription factor SF-1 (steroidogenic factor-1) is a master regulator of steroidogenesis. Previously, we have found that SF-1 induces the differentiation of mesenchymal stem cells into steroidogenic cells. To elucidate the molecular mechanisms of SF-1-mediated functions, we attempted to identify protein components of the SF-1 nuclear protein complex in differentiated cells. SF-1 immunoaffinity chromatography followed by MS/MS analysis was performed, and 24 proteins were identified. Among these proteins, we focused on C/EBPß (CCAAT/enhancer-binding protein ß), which is an essential transcription factor for ovulation and luteinization, as the transcriptional mechanisms of C/EBPß working together with SF-1 are poorly understood. C/EBPß knockdown attenuated cAMP-induced progesterone production in granulosa tumour-derived KGN cells by altering STAR (steroidogenic acute regulatory protein), CYP11A1 (cytochrome P450, family 11, subfamily A, polypeptide 1) and HSD3B2 (hydroxy-δ-5-steroid dehydrogenase, 3ß- and steroid δ-isomerase 2) expression. EMSA and ChIP assays revealed novel C/EBPß-binding sites in the upstream regions of the HSD3B2 and CYP11A1 genes. These interactions were enhanced by cAMP stimulation. Luciferase assays showed that C/EBPß-responsive regions were found in each promoter and C/EBPß is involved in the cAMP-induced transcriptional activity of these genes together with SF-1. These results indicate that C/EBPß is an important mediator of progesterone production by working together with SF-1, especially under tropic hormone-stimulated conditions.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Progesterona/biosíntesis , Factor Esteroidogénico 1/fisiología , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Células Cultivadas , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Regulación de la Expresión Génica , Humanos , Ratones , Fosfoproteínas , Progesterona/genética , Progesterona Reductasa/genética , Espectrometría de Masas en Tándem
8.
Pulm Pharmacol Ther ; 29(1): 24-30, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24462877

RESUMEN

OBJECTIVES: We recently reported that administration of atrial natriuretic peptide during the perioperative period has prophylactic effects with respect to not only cardiovascular but also respiratory complications following pulmonary resection. However, its mechanisms are not well understood. The objective of the present study was to investigate the mechanism of the prophylactic effects of atrial natriuretic peptide in an acute lung injury model. METHODS: For the evaluation of the early phase of pulmonary inflammation, in vitro and in vivo studies using lipopolysaccharide were used. In the in vitro study, the effects of atrial natriuretic peptide on the induction of E-selectin by lipopolysaccharide in human pulmonary artery endothelial cells were evaluated. In the in vivo study, the effects of atrial natriuretic peptide on lipopolysaccharide-induced inflammatory cell infiltration and cytokine levels including tumor necrosis factor-alpha and interleukin-6 in the bronchoalveolar lavage fluid in the lungs of C57/B6 mice were examined. The number of myeloperoxidase-positive staining cells in the tissue sections of the lung of lipopolysaccharide-administered C57/B6 mice was also evaluated. RESULTS: Atrial natriuretic peptide significantly attenuated the up-regulation of E-selectin expression induced by lipopolysaccharide in human pulmonary artery endothelial cells. There were significantly lower cell counts and levels of tumor necrosis factor-alpha and interleukin-6 in the bronchoalveolar lavage fluid of atrial natriuretic peptide-treated mice compared to control mice after lipopolysaccharide injection. In addition, there were significantly fewer myeloperoxidase-positive cells in atrial natriuretic peptide-treated mice than in control mice after lipopolysaccharide injection. CONCLUSIONS: Atrial natriuretic peptide had a protective effect in the lipopolysaccharide-induced acute lung injury model. Atrial natriuretic peptide may be of value in therapeutic strategies aimed at the treatment of acute lung injury such as pneumonia or acute respiratory distress syndrome.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Factor Natriurético Atrial/farmacología , Células Endoteliales/efectos de los fármacos , Pulmón/efectos de los fármacos , Animales , Líquido del Lavado Bronquioalveolar , Modelos Animales de Enfermedad , Selectina E/genética , Células Endoteliales/metabolismo , Humanos , Interleucina-6/metabolismo , Lipopolisacáridos/toxicidad , Pulmón/citología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos
10.
Biochem Pharmacol ; 214: 115663, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37336252

RESUMEN

Fibrosis occurs in all organs and tissues except the brain, and its progression leads to dysfunction of affected organs. Fibrosis-induced organ dysfunction results from the loss of elasticity, strength, and functionality of tissues due to the extracellular matrix secreted by myofibroblasts that express smooth muscle-type actin as a marker. Myofibroblasts, which play a major role in fibrosis, were once thought to originate exclusively from activated fibroblasts; however, it is now clear that myofibroblasts are diverse in origin, from epithelial cells, endothelial cells, adipocytes, macrophages, and other cells. Fibrosis of vital organs, such as the heart, lungs, kidneys, and liver, is a serious chronic disease that ultimately leads to death. Currently, anti-cancer drugs have made remarkable progress, as evidenced by the development of many molecular-targeted drugs, and are making a significant contribution to improving the prognosis of cancer treatment. However, the development of anti-fibrotic agents, which also play an important role in prognosis, has lagged. In this review, the current knowledge regarding myofibroblasts is summarized, with particular attention given to their origin and transdifferentiation signaling pathways (e.g., TGF-ß, Wnt/ß-catenin, YAP/TAZ and AMPK signaling pathways). The development of new small molecule anti-fibrotic agents and the repositioning of existing drugs targeting myofibroblast transdifferentiation are discussed.


Asunto(s)
Transdiferenciación Celular , Miofibroblastos , Humanos , Miofibroblastos/metabolismo , Antifibróticos , Células Endoteliales , Fibroblastos/metabolismo , Fibrosis
11.
Life Sci ; 335: 122278, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37981227

RESUMEN

AIMS: Differentiation-inducing factor-1 (DIF-1), a compound in Dictyostelium discoideum, exhibits anti-cancer effects by inhibiting cell proliferation and motility of various mammalian cancer cells in vitro and in vivo. In addition, DIF-1 suppresses lung colony formation in a mouse model, thus impeding cancer metastasis. However, the precise mechanism underlying its anti-metastatic effect remains unclear. In the present study, we aim to elucidate this mechanism by investigating the adhesion of circulating tumor cells to blood vessels using in vitro and in vivo systems. MAIN METHODS: Melanoma cells (1.0 × 105 cells) were injected into the tail vein of 8-week-old male C57BL/6 mice after administration of DIF-1 (300 mg/kg per day) and/or lipopolysaccharide (LPS: 2.5 mg/kg per day). To investigate cell adhesion and molecular mechanisms, cell adhesion assay, western blotting, immunofluorescence staining, and flow cytometry were performed. KEY FINDINGS: Intragastric administration of DIF-1 suppressed lung colony formation. DIF-1 also substantially inhibited the adhesion of cancer cells to human umbilical vein endothelial cells. Notably, DIF-1 did not affect the expression level of adhesion-related proteins in cancer cells, but it did decrease the expression of vascular cell adhesion molecule-1 (VCAM-1) in human umbilical vein endothelial cells by suppressing its mRNA-to-protein translation through inhibition of mTORC1-p70 S6 kinase signaling. SIGNIFICANCE: DIF-1 reduced tumor cell adhesion to blood vessels by inhibiting mTORC1-S6K signaling and decreasing the expression of adhesion molecule VCAM-1 on vascular endothelial cells. These findings highlight the potential of DIF-1 as a promising compound for the development of anti-cancer drugs with anti-metastatic properties.


Asunto(s)
Dictyostelium , Molécula 1 de Adhesión Celular Vascular , Ratones , Animales , Masculino , Humanos , Molécula 1 de Adhesión Celular Vascular/metabolismo , Lipopolisacáridos/farmacología , Dictyostelium/metabolismo , Ratones Endogámicos C57BL , Proteínas , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Diferenciación Celular , Adhesión Celular , Mamíferos/metabolismo
12.
J Mol Cell Cardiol ; 53(3): 420-8, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22796574

RESUMEN

We have reported that systemic administration of autologous bone marrow or allogenic fetal membrane (FM)-derived mesenchymal stem cells (MSCs) similarly attenuated myocardial injury in rats with experimental autoimmune myocarditis (EAM). Since rat EAM is a T-helper (Th) cell-mediated autoimmune disease, and recent evidence has indicated that both autologous and allogenic MSCs exert an immunosuppressive effect on Th cell activity, we focused on Th cell differentiation in allogenic FM-MSC administered EAM rats. EAM was induced in Lewis rats by injecting porcine cardiac myosin (day 0). Allogenic FM-MSCs, obtained from major histocompatibility complex mismatched ACI rats, were intravenously injected (5 × 10(5)cells/rat) on days 7, 10, or 14 (MSCd7, MSCd10, or MSCd14 groups, respectively). At day 21, echocardiography confirmed that reduced ejection fraction in the untreated EAM group (63 ± 2%) was significantly improved in the MSCd10 and MSCd14 groups (74 ± 1 and 75 ± 2%, respectively, P<0.01). CD68 immunostaining revealed that prominent macrophage infiltration in the myocardium of the EAM group (1466 ± 93 cells/mm(2)) was significantly decreased in the MSCd10 group (958 ± 139 cells/mm(2), P<0.05). To evaluate Th cell differentiation, we used flow cytometry to determine the percentage of interferon (IFN)-γ positive Th1 and interleukin (IL)-17 positive Th17 cells in peripheral CD4-positive Th cells. The percentage of Th1 cells at day 16 was significantly lower in the MSCd10 (1.3 ± 0.2%) and MSCd14 (1.6 ± 0.3%) groups compared to the EAM group (2.4 ± 0.3%, P<0.05), as was the percentage of Th17 cells in the MSCd10 group (1.9 ± 0.5%) compared to the EAM group (2.2 ± 0.9%, P<0.05). At day 21, infiltrating Th17 cells in myocardium were significantly decreased in the MSCd10 group (501 ± 132 cells/mm(2), P<0.05) compared to EAM (921 ± 109 cells/mm(2)). In addition, human CD4+ Th cells co-cultured with human FM-MSCs exhibited reduced Th1 and Th17 cell-differentiation and proliferation, with increased expression of immunosuppressive molecules including indoleamine 2,3-dioxygenase 2 and IL-6 in co-cultured FM-MSCs. These results suggest that intravenous administration of allogenic FM-MSCs ameliorates EAM via the suppression of Th1/Th17 immunity.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Terapia de Inmunosupresión , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Miocarditis/inmunología , Células TH1/inmunología , Células Th17/inmunología , Animales , Enfermedades Autoinmunes/patología , Linfocitos T CD4-Positivos/inmunología , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Ecocardiografía , Corazón/fisiopatología , Interferón gamma/inmunología , Interleucina-17/inmunología , Miocarditis/patología , Miocarditis/terapia , Miocardio/inmunología , Miocardio/patología , Ratas , Ratas Endogámicas ACI , Trasplante Homólogo
13.
Hypertens Res ; 45(12): 1869-1881, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36171325

RESUMEN

Renin-angiotensin system inhibitors have been shown to prevent cancer metastasis in experimental models, but there are limited data in clinical studies. We aimed to explore whether renin-angiotensin system inhibitors administered during the period of cancer resection can influence the subsequent development of metastasis by analyzing multiple individual types of primary cancers. A total of 4927 patients who had undergone resection of primary cancers at Kyushu University Hospital from 2009 to 2014 were enrolled and categorized into 3 groups based on the use of antihypertensive drugs: renin-angiotensin system inhibitors, other drugs, and none. Cumulative incidence functions of metastasis, treating death as a competing risk, were calculated, and the difference was examined among groups by Gray's test. Fine and Gray's model was employed to evaluate multivariate-adjusted hazards of incidental metastasis. In the multivariate-adjusted analysis, patients with skin and renal cancers showed statistically higher risks of metastasis with the use of renin-angiotensin system inhibitors (hazard ratio [95% confidence interval], 5.81 [1.07-31.57] and 4.24 [1.71-10.53], respectively). Regarding pancreatic cancer, patients treated with antihypertensive drugs other than renin-angiotensin system inhibitors had a significantly increased risk of metastasis (hazard ratio [95% confidence interval], 3.31 [1.43-7.69]). Future larger studies are needed to ascertain whether renin-angiotensin system inhibitors can increase the risk of metastasis in skin and renal cancers, focusing on specific tissue types and potential factors associated with renin-angiotensin system inhibitor use.


Asunto(s)
Neoplasias Renales , Neoplasias Pancreáticas , Humanos , Antihipertensivos/uso terapéutico , Antagonistas de Receptores de Angiotensina/uso terapéutico , Antagonistas de Receptores de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Sistema Renina-Angiotensina , Estudios Retrospectivos , Registros Electrónicos de Salud , Inhibidores Enzimáticos/farmacología , Neoplasias Pancreáticas/inducido químicamente , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Renales/tratamiento farmacológico
14.
Biochem Pharmacol ; 197: 114950, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35143754

RESUMEN

We previously reported that 2,5-dimethylcelecoxib (DM-C), a derivative of celecoxib, lacks cyclooxygenase-2 inhibitory effects and suppresses cardiac remodeling by activating glycogen synthase kinase-3 (GSK-3). However, it remains unclear whether DM-C attenuates fibroblast-to-myofibroblast transformation (FMT), which plays a key role in cardiac fibrosis. Therefore, we evaluated the effect of DM-C on FMT using a cryoinjury-induced myocardial infarction (CMI) mouse model. We found that DM-C attenuated the deterioration of left ventricular ejection fraction after CMI by decreasing cardiac fibrosis. Analysis of the expression level of α-smooth muscle actin (α-SMA), a marker for myofibroblasts, indicated that DM-C decreased FMT at the cardiac injury site. To investigate the mechanism by which DM-C attenuated FMT, fibroblasts obtained from the heart were stimulated with TGF-ß to induce FMT, and the effect of DM-C was analyzed. DM-C suppressed the expression of α-SMA and the phosphorylation levels of Smad 2/3 and GSK-3, indicating that DM-C suppressed α-SMA expression by inhibiting the transforming growth factor (TGF)-ß signaling pathway via activation of GSK-3. DM-C decreased the expression of collagen, connective tissue growth factor (CTGF) and Snail, which are also known to accelerate cardiac fibrosis. These results suggested that DM-C attenuated cardiac fibrosis by suppressing FMT at the injured site after CMI by inhibiting the TGF-ß signaling pathway via activation of GSK-3. Thus, DM-C has potential against cardiac disease as a novel anti-fibrotic agent.


Asunto(s)
Fibroblastos/efectos de los fármacos , Congelación/efectos adversos , Infarto del Miocardio/tratamiento farmacológico , Miofibroblastos/efectos de los fármacos , Pirazoles/uso terapéutico , Transducción de Señal/efectos de los fármacos , Sulfonamidas/uso terapéutico , Animales , Células Cultivadas , Fibroblastos/enzimología , Fibroblastos/patología , Fibrosis , Glucógeno Sintasa Quinasa 3/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/enzimología , Infarto del Miocardio/etiología , Infarto del Miocardio/patología , Miofibroblastos/enzimología , Miofibroblastos/patología , Nitrógeno/toxicidad , Pirazoles/farmacología , Ratas , Ratas Endogámicas Lew , Transducción de Señal/fisiología , Sulfonamidas/farmacología
15.
Biochim Biophys Acta Mol Cell Res ; 1868(2): 118909, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33189784

RESUMEN

Mesenchymal stem cells (MSCs) are an attractive cell source for tissue regeneration and repair. However, their low differentiation efficacy currently impedes the development of MSC therapy. Therefore, in this study, we investigated the effects of differentiation-inducing factor-1 (DIF-1) on the differentiation efficacy of bone marrow-derived MSCs (BM-MSCs) into adipogenic or osteogenic lineages. BM-MSCs, which were obtained from Sprague-Dawley rats, were positive for the MSC markers (CD29, CD73, and CD90). DIF-1 alone neither affected cell surface antigen expression nor induced adipogenic or osteogenic differentiation. However, DIF-1 significantly enhanced the effects of adipogenic differentiation stimuli, which were evaluated as the number of oil red-O positive cells and the expression of adipocyte differentiation markers (peroxisome proliferator-activated receptor gamma, adipocyte fatty acid-binding protein, and adiponectin). In contrast, DIF-1 significantly attenuated the effects of osteogenic differentiation stimuli, which were evaluated as alizarin red-S positive calcium deposition, and the expression of osteoblast differentiation markers alkaline phosphatase, runt-related transcription factor 2, and osteopontin. We further investigated the mechanism by which DIF-1 affects MSC differentiation efficacy and found that glycogen synthase kinase-3 was the main factor mediating the action of DIF-1 on the adipogenic differentiation of BM-MSCs, whereas it was only partially involved in osteogenic differentiation. These results suggest that DIF-1 supports MSC differentiation toward the desired cell fate by enhancing the differentiation efficacy.


Asunto(s)
Adipogénesis/efectos de los fármacos , Hexanonas/farmacología , Hidrocarburos Clorados/farmacología , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/efectos de los fármacos , Adipocitos/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Glucógeno Sintasa Quinasa 3/metabolismo , Masculino , Osteoblastos/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
16.
Eur J Pharmacol ; 909: 174415, 2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34375673

RESUMEN

Angiotensin II (Ang II) reportedly facilitates primary tumor growth and distal hematogenous metastasis formation in various murine intravenous metastasis models. However, it is unclear whether Ang II accelerates the initial processes of metastasis formation that begins in primary tumors surrounded by tumor microenvironment. We examined the effects of Ang II on primary tumors and lung metastasis lesions using a murine spontaneous metastasis model, in which triple negative breast cancer 4T1 cells constitutively expressing luciferase (4T1-Luc cells) were injected into the mammary fat pad of BALB/c mice. Subcutaneous injection of Ang II significantly accelerated primary tumor growth and lung metastasis formation. Ang II increased the protein expression levels of c-Myc, cyclin D1, fibronectin, vimentin, αSMA and Snail, and the treatment with the Ang II type 1 receptor blocker valsartan significantly suppressed the Ang II-induced increases of fibronectin and vimentin. Valsartan also significantly reduced lung metastatic lesions. However, Ang II did not have significant effects on 4T1-Luc cells including the proliferation, migration, invasion, or the expressions of proteins related to cell proliferation and epithelial-to-mesenchymal transition. In contrast, when 4T1-Luc cells were co-cultured with dermal fibroblasts, Ang II significantly accelerated cell migration and increased the expressions of fibronectin, vimentin, αSMA and Snail in 4T1-Luc cells. And moreover, Ang II significantly increased the mRNA expression of IL-6 in fibroblasts co-cultured with 4T1-Luc cells. These results suggested that Ang II accelerates surrounding fibroblasts by soluble factors such as IL-6 to promote epithelial-to-mesenchymal transition, which result in the initiation of cancer metastasis.


Asunto(s)
Angiotensina II/metabolismo , Fibroblastos Asociados al Cáncer/patología , Neoplasias Pulmonares/secundario , Neoplasias de la Mama Triple Negativas/patología , Animales , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Humanos , Pulmón/patología , Glándulas Mamarias Animales/patología , Ratones , Microambiente Tumoral
17.
J Mol Cell Cardiol ; 49(5): 753-61, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20692268

RESUMEN

We reported previously that the autologous administration of bone marrow-derived mesenchymal stem cells (BM-MSC) significantly attenuated myocardial dysfunction and injury in a rat model of acute myocarditis by stimulating angiogenesis and reducing inflammation. Because BM aspiration procedures are invasive and can yield low numbers of MSC after processing, we focused on fetal membranes (FMs) as an alternative source of MSC to provide a large number of cells. We investigated whether the allogeneic administration of FM-derived MSC (FM-MSC) attenuates myocardial injury and dysfunction in a rat myocarditis model. Experimental autoimmune myocarditis (EAM) was induced in male Lewis rats by injecting porcine cardiac myosin. Allogeneic FM-MSC obtained from major histocompatibility complex-mismatched ACI rats (5 × 10(5) cells/animal) were injected intravenously into Lewis rats one week after myosin administration. At day 21, severe cardiac inflammation and deterioration of cardiac function were observed. The allogeneic administration of FM-MSC significantly attenuated inflammatory cell infiltration and monocyte chemoattractant protein 1 expression in the myocardium and improved cardiac function. In a T-lymphocyte proliferation assay, the proliferative response of splenic T lymphocytes was significantly lower in cells obtained from FM-MSC-treated EAM rats that reacted to myosin than in cells obtained from vehicle-treated rats with EAM. T-lymphocyte activation was significantly reduced by coculture with FM-MSC. The allogeneic administration of FM-MSC attenuated myocardial dysfunction and inflammation, and the host cell-mediated immune response was attenuated in a rat model of acute myocarditis. These results suggest that allogeneic administration of FM-MSC might provide a new therapeutic strategy for the treatment of acute myocarditis.


Asunto(s)
Membranas Extraembrionarias/citología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Miocarditis/terapia , Enfermedad Aguda , Animales , Proliferación Celular , Pruebas de Función Cardíaca , Inflamación/complicaciones , Inflamación/patología , Inyecciones Intravenosas , Activación de Linfocitos/inmunología , Miocarditis/diagnóstico por imagen , Miocarditis/patología , Miocarditis/fisiopatología , Miocardio/patología , Ratas , Linfocitos T/citología , Linfocitos T/inmunología , Trasplante Homólogo , Ultrasonografía
18.
Am J Physiol Renal Physiol ; 299(5): F1004-13, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20739390

RESUMEN

Mesenchymal stem cells (MSC) have been reported to be an attractive therapeutic cell source for the treatment of renal diseases. Recently, we reported that transplantation of allogenic fetal membrane-derived MSC (FM-MSC), which are available noninvasively in large amounts, had a therapeutic effect on a hindlimb ischemia model (Ishikane S, Ohnishi S, Yamahara K, Sada M, Harada K, Mishima K, Iwasaki K, Fujiwara M, Kitamura S, Nagaya N, Ikeda T. Stem Cells 26: 2625-2633, 2008). Here, we investigated whether allogenic FM-MSC administration could ameliorate renal injury in experimental glomerulonephritis. Lewis rats with anti-Thy1 nephritis intravenously received FM-MSC obtained from major histocompatibility complex-mismatched ACI rats (FM-MSC group) or a PBS (PBS group). Nephritic rats exhibited an increased urinary protein excretion in the PBS group, whereas the FM-MSC group rats had a significantly lower level of increase (P < 0.05 vs. PBS group). FM-MSC transplantation significantly reduced activated mesangial cell (MC) proliferation, glomerular monocyte/macrophage infiltration, mesangial matrix accumulation, as well as the glomerular expression of inflammatory or extracellular matrix-related genes including TNF-α, monocyte chemoattractant protein 1 (MCP-1), type I collagen, TGF-ß, type 1 plasminogen activator inhibitor (PAI-1) (P < 0.05 vs. PBS group). In vitro, FM-MSC-derived conditioned medium significantly attenuated the expression of TNF-α and MCP-1 in rat MC through a prostaglandin E(2)-dependent mechanism. These data suggest that transplanted FM-MSC contributed to the healing process in injured kidney tissue by producing paracrine factors. Our results indicate that allogenic FM-MSC transplantation is a potent therapeutic strategy for the treatment of acute glomerulonephritis.


Asunto(s)
Membranas Extraembrionarias/citología , Glomerulonefritis/terapia , Trasplante de Células Madre Mesenquimatosas , Actinas/metabolismo , Animales , Western Blotting , Proliferación Celular , Quimiocinas/biosíntesis , Medios de Cultivo Condicionados , Citocinas/biosíntesis , Dinoprostona/metabolismo , Ensayo de Inmunoadsorción Enzimática , Mesangio Glomerular/citología , Mesangio Glomerular/fisiología , Glomerulonefritis/inducido químicamente , Glomerulonefritis/patología , Inmunohistoquímica , Riñón/citología , Riñón/patología , Células Mesangiales/fisiología , Monocitos/fisiología , Comunicación Paracrina/fisiología , Proteinuria/terapia , Ratas , Ratas Endogámicas Lew , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/biosíntesis
19.
Stem Cells ; 26(10): 2625-33, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18669910

RESUMEN

Bone marrow-derived mesenchymal stem cells (BM-MSC) have been demonstrated to be an attractive therapeutic cell source for tissue regeneration and repair. However, it remains unknown whether or not allogeneic transplantation of mesenchymal stem cells (MSC) derived from fetal membranes (FM), which are generally discarded as medical waste after delivery, has therapeutic potential. FM-MSC were obtained from Lewis rats and had surface antigen expression and multipotent potential partly similar to those of BM-MSC. Compared with BM-MSC, FM-MSC secreted a comparable amount of hepatocyte growth factor despite a small amount of vascular endothelial growth factor. FM-MSC and BM-MSC both expressed major histocompatibility complex (MHC) class I but not MHC class II antigens and did not elicit allogeneic lymphocyte proliferation in mixed lymphocyte culture. FM-MSC or BM-MSC obtained from Lewis rats were injected into a MHC-mismatched August-Copenhagen-Irish rat model of hind limb ischemia. Three weeks after injection, blood perfusion and capillary density were significantly higher in the FM-MSC and BM-MSC groups than in the phosphate-buffered saline group, and allogeneic FM-MSC and BM-MSC were still observed. In nonischemic hind limb tissues, allogeneic FM-MSC and BM-MSC injection were associated with a comparatively small amount of T lymphocyte infiltration, compared with the injection of allogeneic splenic lymphocytes. In conclusion, allogeneic FM-MSC injection did not elicit a lymphocyte proliferative response and provided significant improvement in a rat model of hind limb ischemia, comparable to the response to BM-MSC. Thus, allogeneic injection of FM-MSC may be a new therapeutic strategy for the treatment of severe peripheral vascular disease. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Membranas Extraembrionarias/citología , Miembro Posterior/irrigación sanguínea , Miembro Posterior/patología , Isquemia/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica , Inductores de la Angiogénesis/metabolismo , Animales , Células de la Médula Ósea/citología , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Inyecciones , Prueba de Cultivo Mixto de Linfocitos , Masculino , Músculos/citología , Músculos/inmunología , Ratas , Ratas Endogámicas Lew , Linfocitos T/citología , Trasplante Homólogo
20.
Biol Pharm Bull ; 32(9): 1538-44, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19721229

RESUMEN

Cannabidiol decreases cerebral infarction and high-mobility group box1 (HMGB1) in plasma in ischemic early phase. However, plasma HMGB1 levels in ischemic delayed phase reach higher concentration with the progressing brain injury. In this study, we investigated the therapeutic time window of cannabidiol on functional deficits, glial HMGB1 and plasma HMGB1 levels in a 4 h mouse middle cerebral artery (MCA) occlusion model. Cannabidiol-treated mice were divided into 3 groups as follows: group (a) treated from day 1, group (b) treated from day 3, group (c) treated from day 5 after MCA occlusion. Moreover, minocycline, microglia inhibitor, and fluorocitrate, an inhibitor of astroglial metabolism, were used to compare with cannabidiol-treated group. Repeated treatment with cannabidiol from 1 and 3 d at the latest after cerebral ischemia improved functional deficits and survival rates. However, cannabidiol from 5 d could not improve the ischemic damage as well as fluorocitrate-treated group. Moreover, both group (a), group (b) and minocycline but not group (c) and fluorocitrate-treated group had a decrease in the number of Iba1 expressing HMGB1 positive cells and HMGB1 levels in plasma. Cannabidiol may provide therapeutic possibilities for the progressing brain injury via HMGB1-inhibiting mechanism.


Asunto(s)
Isquemia Encefálica/sangre , Isquemia Encefálica/tratamiento farmacológico , Cannabidiol/uso terapéutico , Proteína HMGB1/sangre , Animales , Biomarcadores/sangre , Isquemia Encefálica/patología , Masculino , Ratones , Factores de Tiempo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA