Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Asian Pac J Cancer Prev ; 23(2): 601-615, 2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-35225473

RESUMEN

INTRODUCTION: There are limited published data regarding the recent incidence trends of cancer in Iraqi Kurdistan. METHODS: The present study assessed the epidemiological estimates of cancer incidence, as well providing a projection of future cancer trends in the upcoming decade by analysing the population-based cancer registry between 2013 and 2019, in both the Erbil and Duhok governorates. A retrospective analysis was performed on data retrieved from the Medical Statistics Department at the Ministry of Health, Kurdistan Regional Government (KRG). RESULTS: The total number of female cancer patients was higher in both governorates, and the total incidence of patients with cancer increased by over 2x between 2013 and 2019 in Erbil and Duhok, from 73 to 174 patients/100,000 individuals for women, and 36 to 85 patients/100,000 individuals for men. Analysis indicated that the percentage of patients with cancer is projected to increase by >2x in the current decade, from 3,457 cases to 4,547 and 4,449 cases in the Erbil governorate; and from 1,365 to 2,633 and 2,737 cases in 2028 based on LSTM and bi-LTSM analysis in the Duhok governorate. Lung cancer (LC) and female breast cancer (BC) were the most prominent types of cancers diagnosed since 2013 in both the Erbil and Duhok governorates. CONCLUSION: The striking pattern of trends for both present and future cancer incidence rates require urgent solutions and comprehensive efforts to control risk factors that promote the increasing incidence of cancer in these two KRG governorates.
.


Asunto(s)
Neoplasias/epidemiología , Adolescente , Adulto , Distribución por Edad , Anciano , Niño , Preescolar , Femenino , Humanos , Incidencia , Lactante , Recién Nacido , Irak/epidemiología , Masculino , Persona de Mediana Edad , Sistema de Registros , Estudios Retrospectivos , Distribución por Sexo , Adulto Joven
2.
Circulation ; 136(5): 464-475, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28473446

RESUMEN

BACKGROUND: Thioredoxin (TRX)-1, a ubiquitous 12-kDa protein, exerts antioxidant and anti-inflammatory effects. In contrast, the truncated form, called TRX80, produced by macrophages induces upregulation of proinflammatory cytokines. TRX80 also promotes the differentiation of mouse peritoneal and human macrophages toward a proinflammatory M1 phenotype. METHODS: TRX1 and TRX80 plasma levels were determined with a specific ELISA. A disintegrin and metalloproteinase domain-containing protein (ADAM)-10, ADAM-17, and ADAM-10 activities were measured with SensoLyte 520 ADAM10 Activity Assay Kit, Fluorimetric, and InnoZyme TACE Activity Kit, respectively. Western immunoblots were performed with specific antibodies to ADAM-10 or ADAM-17. Angiogenesis study was evaluated in vitro with human microvascular endothelial cells-1 and in vivo with the Matrigel plug angiogenesis assay in mice. The expression of macrophage phenotype markers was investigated with real-time polymerase chain reaction. Phosphorylation of Akt, mechanistic target of rapamycin, and 70S6K was determined with specific antibodies. The effect of TRX80 on NLRP3 inflammasome activity was evaluated by measuring the level of interleukin-1ß and -18 in the supernatants of activated macrophages with ELISA. Hearts were used for lesion surface evaluation and immunohistochemical studies, and whole descending aorta were stained with Oil Red O. For transgenic mice generation, the human scavenger receptor (SR-A) promoter/enhancer was used to drive macrophage-specific expression of human TRX80 in mice. RESULTS: In this study, we observed a significant increase of plasma levels of TRX80 in old subjects compared with healthy young subjects. In parallel, an increase in expression and activity of ADAM-10 and ADAM-17 in old peripheral blood mononuclear cells compared with those of young subjects was observed. Furthermore, TRX80 was found to colocalize with tumor necrosis factor-α, a macrophage M1 marker, in human atherosclerotic plaque. In addition, TRX80 induced the expression of murine M1 macrophage markers through Akt2/mechanistic target of rapamycin-C1/70S6K pathway and activated the inflammasome NLRP3, leading to the release of interleukin-1ß and -18, potent atherogenic cytokines. Moreover, TRX80 exerts a powerful angiogenic effect in both in vitro and in vivo mouse studies. Finally, transgenic mice that overexpress human TRX80 specifically in macrophages of apoE-/- mice have a significant increase of aortic atherosclerotic lesions. CONCLUSIONS: TRX80 showed an age-dependent increase in human plasma. In mouse models, TRX80 was associated with a proinflammatory status and increased atherosclerosis.


Asunto(s)
Envejecimiento , Aterosclerosis/patología , Fragmentos de Péptidos/sangre , Tiorredoxinas/sangre , Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Adulto , Anciano , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/metabolismo , Biomarcadores/sangre , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunohistoquímica , Inflamación , Interleucina-18/sangre , Interleucina-1beta/sangre , Leucocitos Mononucleares/metabolismo , Lipopolisacáridos/toxicidad , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Tiorredoxinas/farmacología
4.
Mol Nutr Food Res ; 59(5): 939-47, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25644174

RESUMEN

SCOPE: Formula-derived dietary advanced glycation end products (AGEs) may promote programming of inflammation and oxidative stress in the kidney of intrauterine growth retardation (IUGR) piglets. METHODS AND RESULTS: IUGR piglets received either a low temperature heated formula (n = 8) or a high temperature heated formula (HHF: n = 8) or suckled naturally for 3 wk postnatally. Then they were fed with normal ad libitum regular diet. N(ε)-carboxymethyllysine (CML) was measured in plasma, feces, and formula by HPLC/MS-MS. CML was detected by immunofluorescence in kidney cells. Target renin-angiotensin-apoptotic, pro-inflammatory genes-p62 NF-κB, and soluble receptor of AGE (sRAGE) levels were quantified. Compared with that in controls, free CML and plasma urea increased significantly in the HHF-fed group at PND36 (p < 0.05). CML was detected in the nuclei of renal tubular cells of formula-fed piglets but not in suckled ones. This presence of CML was associated with the activation of the soluble receptor of AGE. AT1, AT2, caspase 3, caspase 8, NF-κB, p62 NF-κB, and total protein oxidation in kidney were higher in HHF-fed group as compared to LHF-fed group (p < 0.05). CONCLUSION: Food processes aimed at reducing the concentration of AGEs in infant formula are urgently needed and may be therapeutically relevant for premature and/or IUGR babies.


Asunto(s)
Retardo del Crecimiento Fetal/metabolismo , Productos Finales de Glicación Avanzada/administración & dosificación , Inflamación/etiología , Riñón/metabolismo , Estrés Oxidativo , Animales , Núcleo Celular/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Riñón/crecimiento & desarrollo , Peroxidación de Lípido , Hígado/metabolismo , Lisina/análogos & derivados , Lisina/metabolismo , Receptor para Productos Finales de Glicación Avanzada/análisis , Sistema Renina-Angiotensina/fisiología , Porcinos
5.
Circulation ; 131(12): 1061-70, 2015 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-25613820

RESUMEN

BACKGROUND: This study was designed to evaluate the effect of arglabin on the NLRP3 inflammasome inhibition and atherosclerotic lesion in ApoE2Ki mice fed a high-fat Western-type diet. METHODS AND RESULTS: Arglabin was purified, and its chemical identity was confirmed by mass spectrometry. It inhibited, in a concentration-dependent manner, interleukin (IL)-1ß and IL-18, but not IL-6 and IL-12, production in lipopolysaccharide and cholesterol crystal-activated cultured mouse peritoneal macrophages, with a maximum effect at ≈50 nmol/L and EC50 values for both cytokines of ≈ 10 nmol/L. Lipopolysaccharide and cholesterol crystals did not induce IL-1ß and IL-18 production in Nlrp3(-/-) macrophages. In addition, arglabin activated autophagy as evidenced by the increase in LC3-II protein. Intraperitoneal injection of arglabin (2.5 ng/g body weight twice daily for 13 weeks) into female ApoE2.Ki mice fed a high-fat diet resulted in a decreased IL-1ß plasma level compared with vehicle-treated mice (5.2±1.0 versus 11.7±1.1 pg/mL). Surprisingly, arglabin also reduced plasma levels of total cholesterol and triglycerides to 41% and 42%, respectively. Moreover, arglabin oriented the proinflammatory M1 macrophages into the anti-inflammatory M2 phenotype in spleen and arterial lesions. Finally, arglabin treatment markedly reduced the median lesion areas in the sinus and whole aorta to 54% (P=0.02) and 41% (P=0.02), respectively. CONCLUSIONS: Arglabin reduces inflammation and plasma lipids, increases autophagy, and orients tissue macrophages into an anti-inflammatory phenotype in ApoE2.Ki mice fed a high-fat diet. Consequently, a marked reduction in atherosclerotic lesions was observed. Thus, arglabin may represent a promising new drug to treat inflammation and atherosclerosis.


Asunto(s)
Apolipoproteína E2/deficiencia , Aterosclerosis/tratamiento farmacológico , Proteínas Portadoras/antagonistas & inhibidores , Dieta Alta en Grasa/efectos adversos , Inflamasomas/antagonistas & inhibidores , Sesquiterpenos/uso terapéutico , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Aterosclerosis/sangre , Aterosclerosis/etiología , Femenino , Inflamasomas/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Sesquiterpenos/farmacología , Sesquiterpenos de Guayano , Resultado del Tratamiento
6.
J Cell Physiol ; 228(7): 1577-83, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23335265

RESUMEN

Vascular cells are particularly susceptible to oxidative stress that is believed to play a key role in the pathogenesis of cardiovascular disorders. Thioredoxin-1 (Trx-1) is an oxidative stress-limiting protein with anti-inflammatory and anti-apoptotic properties. In contrast, its truncated form (Trx-80) exerts pro-inflammatory effects. Here we analyzed whether Trx-80 might exert atherogenic effects by promoting macrophage differentiation into the M1 pro-inflammatory phenotype. Trx-80 at 1 µg/ml significantly attenuated the polarization of anti-inflammatory M2 macrophages induced by exposure to either IL-4 at 15 ng/ml or IL-4/IL-13 (10 ng/ml each) in vitro, as evidenced by the expression of the characteristic markers, CD206 and IL-10. By contrast, in LPS-challenged macrophages, Trx-80 significantly potentiated the differentiation into inflammatory M1 macrophages as indicated by the expression of the M1 cytokines, TNF-α and MCP-1. When Trx-80 was administered to hyperlipoproteinemic ApoE2.Ki mice at 30 µg/g body weight (b.w.) challenged either with LPS at 30 µg/30 g (b.w.) or IL-4 at 500 ng/30 g (b.w.), it significantly induced the M1 phenotype but inhibited differentiation of M2 macrophages in thymus and liver. When ApoE2.Ki mice were challenged once weekly with LPS for 5 weeks, they showed severe atherosclerotic lesions enriched with macrophages expressing predominantly M1 over M2 markers. Such effect was potentiated when mice received daily, in addition to LPS, the Trx-80. Moreover, the Trx-80 treatment led to a significantly increased aortic lesion area. The ability of Trx-80 to promote differentiation of macrophages into the classical proinflammatory phenotype may explain its atherogenic effects in cardiovascular diseases.


Asunto(s)
Aterosclerosis/fisiopatología , Inflamación/fisiopatología , Macrófagos/fisiología , Fragmentos de Péptidos/fisiología , Tiorredoxinas/fisiología , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Apolipoproteína E2/genética , Apolipoproteína E2/metabolismo , Aterosclerosis/etiología , Aterosclerosis/patología , Biomarcadores/metabolismo , Diferenciación Celular , Humanos , Inflamación/etiología , Inflamación/patología , Lectinas Tipo C/metabolismo , Macrófagos/clasificación , Macrófagos/patología , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Fenotipo , Receptores de Superficie Celular/metabolismo
7.
Antioxid Redox Signal ; 19(11): 1266-303, 2013 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-23244617

RESUMEN

The thioredoxin (Trx) system comprises Trx, truncated Trx (Trx-80), Trx reductase, and NADPH, besides a natural Trx inhibitor, the thioredoxin-interacting protein (TXNIP). This system is essential for maintaining the balance of the cellular redox status, and it is involved in the regulation of redox signaling. It is also pivotal for growth promotion, neuroprotection, inflammatory modulation, antiapoptosis, immune function, and atherosclerosis. As an ubiquitous and multifunctional protein, Trx is expressed in all forms of life, executing its function through its antioxidative, protein-reducing, and signal-transducing activities. In this review, the biological properties of the Trx system are highlighted, and its implications in several human diseases are discussed, including cardiovascular diseases, heart failure, stroke, inflammation, metabolic syndrome, neurodegenerative diseases, arthritis, and cancer. The last chapter addresses the emerging therapeutic approaches targeting the Trx system in human diseases.


Asunto(s)
Tiorredoxinas/metabolismo , Envejecimiento , Animales , Enfermedades Cardiovasculares/metabolismo , Ejercicio Físico , Humanos , Inmunidad , Inflamación/metabolismo , Enfermedades Metabólicas/metabolismo , Neoplasias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Reductasa de Tiorredoxina-Disulfuro/antagonistas & inhibidores , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Tiorredoxinas/antagonistas & inhibidores
8.
Arterioscler Thromb Vasc Biol ; 32(6): 1445-52, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22516068

RESUMEN

OBJECTIVE: Oxidative stress is believed to play a key role in cardiovascular disorders. Thioredoxin (Trx) is an oxidative stress-limiting protein with anti-inflammatory and antiapoptotic properties. Here, we analyzed whether Trx-1 might exert atheroprotective effects by promoting macrophage differentiation into the M2 anti-inflammatory phenotype. METHODS AND RESULTS: Trx-1 at 1 µg/mL induced downregulation of p16(INK4a) and significantly promoted the polarization of anti-inflammatory M2 macrophages in macrophages exposed to interleukin (IL)-4 at 15 ng/mL or IL-4/IL-13 (10 ng/mL each) in vitro, as evidenced by the expression of the CD206 and IL-10 markers. In addition, Trx-1 induced downregulation of nuclear translocation of activator protein-1 and Ref-1, and significantly reduced the lipopolysaccharide-induced differentiation of inflammatory M1 macrophages, as indicated by the decreased expression of the M1 cytokines, tumor necrosis factor-α and monocyte chemoattractant protein-1. Consistently, Trx-1 administered to hyperlipoproteinemic ApoE2.Ki mice at 30 µg/30 g body weight challenged either with lipopolysaccharide at 30 µg/30 g body weight or with IL-4 at 500 ng/30 g body weight significantly induced the M2 phenotype while inhibiting differentiation of macrophages into the M1 phenotype in liver and thymus. ApoE2.Ki mice challenged once weekly with lipopolysaccharide for 5 weeks developed severe atherosclerotic lesions enriched with macrophages expressing predominantly M1 over M2 markers. In contrast, however, daily injections of Trx-1 shifted the phenotype pattern of lesional macrophages in these animals to predominantly M2 over M1, and the aortic lesion area was significantly reduced (from 100%±18% to 62.8%±9.8%; n=8; P<0.01). Consistently, Trx-1 colocalized with M2 but not with M1 macrophage markers in human atherosclerotic vessel specimens. CONCLUSIONS: The ability of Trx-1 to promote differentiation of macrophages into an alternative, anti-inflammatory phenotype may explain its protective effects in cardiovascular diseases. These data provide novel insight into the link between oxidative stress and cardiovascular diseases.


Asunto(s)
Antiinflamatorios/farmacología , Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Diferenciación Celular/efectos de los fármacos , Macrófagos Peritoneales/efectos de los fármacos , Tiorredoxinas/farmacología , Animales , Enfermedades de la Aorta/inducido químicamente , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apolipoproteína E2/genética , Apolipoproteína E2/metabolismo , Aterosclerosis/inducido químicamente , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Biomarcadores/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Citocinas/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/metabolismo , Lectinas Tipo C/metabolismo , Lipopolisacáridos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fenotipo , Receptores de Superficie Celular/metabolismo , Proteínas Recombinantes/farmacología , Factores de Tiempo , Factor de Transcripción AP-1/metabolismo
9.
J Biol Chem ; 286(33): 28858-28866, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21700710

RESUMEN

Macrophages play a pivotal role in the pathophysiology of atherosclerosis. These cells express cathepsin L (CatL), a cysteine protease that has been implicated in atherogenesis and the associated arterial remodeling. In addition, macrophages highly express peroxisome proliferator-activated receptor (PPAR) γ, a transcription factor that regulates numerous genes important for lipid and lipoprotein metabolism, for glucose homeostasis, and inflammation. Hence, PPARγ might affect macrophage function in the context of chronic inflammation such as atherogenesis. In the present study, we examined the effect of PPARγ activation on the expression of CatL in human monocyte-derived macrophages (HMDM). Activation of PPARγ by the specific agonist GW929 concentration-dependently increased the levels of CatL mRNA and protein in HMDM. By promoter analysis, we identified a functional PPAR response element-like sequence that positively regulates CatL expression. In addition, we found that PPARγ-induced CatL promotes the degradation of Bcl2 without affecting Bax protein levels. Consistently, degradation of Bcl2 could be prevented by a specific CatL inhibitor, confirming the causative role of CatL. PPARγ-induced CatL was found to decrease autophagy through reduction of beclin 1 and LC3 protein levels. The reduction of these proteins involved in autophagic cell death was antagonized either by the CatL inhibitor or by CatL knockdown. In conclusion, our data show that PPARγ can specifically induce CatL, a proatherogenic protease, in HMDM. In turn, CatL inhibits autophagy and induces apoptosis. Thus, the proatherogenic effect of CatL could be neutralized by apoptosis, a beneficial phenomenon, at least in the early stages of atherosclerosis.


Asunto(s)
Apoptosis , Aterosclerosis/metabolismo , Autofagia , Catepsina L/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo , PPAR gamma/metabolismo , Aterosclerosis/genética , Aterosclerosis/patología , Catepsina L/genética , Humanos , Macrófagos/patología , Monocitos/patología , PPAR gamma/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Elementos de Respuesta/genética , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA