Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Biol Chem ; 290(8): 5065-5077, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25548278

RESUMEN

Supplemental oxygen inhalation is frequently used to treat severe respiratory failure; however, prolonged exposure to hyperoxia causes hyperoxic acute lung injury (HALI), which induces acute respiratory distress syndrome and leads to high mortality rates. Recent investigations suggest the possible role of NLRP3 inflammasomes, which regulate IL-1ß production and lead to inflammatory responses, in the pathophysiology of HALI; however, their role is not fully understood. In this study, we investigated the role of NLRP3 inflammasomes in mice with HALI. Under hyperoxic conditions, NLRP3(-/-) mice died at a higher rate compared with wild-type and IL-1ß(-/-) mice, and there was no difference in IL-1ß production in their lungs. Under hyperoxic conditions, the lungs of NLRP3(-/-) mice exhibited reduced inflammatory responses, such as inflammatory cell infiltration and cytokine expression, as well as increased and decreased expression of MMP-9 and Bcl-2, respectively. NLRP3(-/-) mice exhibited diminished expression and activation of Stat3, which regulates MMP-9 and Bcl-2, in addition to increased numbers of apoptotic alveolar epithelial cells. In vitro experiments revealed that alveolar macrophages and neutrophils promoted Stat3 activation in alveolar epithelial cells. Furthermore, NLRP3 deficiency impaired the migration of neutrophils and chemokine expression by macrophages. These findings demonstrate that NLRP3 regulates Stat3 signaling in alveolar epithelial cells by affecting macrophage and neutrophil function independent of IL-1ß production and contributes to the pathophysiology of HALI.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Proteínas Portadoras/genética , Hiperoxia/metabolismo , Interleucina-1beta/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Animales , Proteínas Portadoras/metabolismo , Hiperoxia/genética , Hiperoxia/patología , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología
2.
Am J Physiol Endocrinol Metab ; 311(5): E881-E890, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27702746

RESUMEN

Caspase-1 is a cysteine protease responsible for the processing of the proinflammatory cytokine interleukin-1ß and activated by the formation of inflammasome complexes. Although several investigations have found a link between diet-induced obesity and caspase-1, the relationship remains controversial. Here, we found that mice deficient in caspase-1 were susceptible to high-fat diet-induced obesity with increased adiposity as well as normal lipid and glucose metabolism. Caspase-1 deficiency clearly promoted the infiltration of inflammatory macrophages and increased the production of C-C motif chemokine ligand 2 (CCL2) in the adipose tissue. The dominant cellular source of CCL2 was stromal vascular fraction rather than adipocytes in the adipose tissue. These findings demonstrate a critical role of caspase-1 in macrophage-driven inflammation in the adipose tissue and the development of obesity. These data provide novel insights into the mechanisms underlying inflammation in the pathophysiology of obesity.


Asunto(s)
Tejido Adiposo/inmunología , Caspasa 1/genética , Quimiocina CCL2/inmunología , Macrófagos/inmunología , Obesidad/genética , Adipocitos/inmunología , Adipocitos/patología , Adiponectina/inmunología , Tejido Adiposo/patología , Animales , Glucemia/metabolismo , Composición Corporal , Caspasa 1/inmunología , Colesterol/metabolismo , Dieta Alta en Grasa , Citometría de Flujo , Perfilación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Insulina/metabolismo , Interferón gamma/inmunología , Interleucina-12/inmunología , Interleucina-18/inmunología , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Leptina/inmunología , Masculino , Ratones , Ratones Noqueados , Obesidad/inmunología , Obesidad/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Triglicéridos/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Microtomografía por Rayos X
3.
Arterioscler Thromb Vasc Biol ; 35(1): 127-36, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25378412

RESUMEN

OBJECTIVE: Abdominal aortic aneurysm (AAA) is considered a chronic inflammatory disease; however, the molecular basis underlying the sterile inflammatory response involved in the process of AAA remains unclear. We previously showed that the inflammasome, which regulates the caspase-1-dependent interleukin-1ß production, mediates the sterile cardiovascular inflammatory responses. Therefore, we hypothesized that the inflammasome is a key mediator of initial inflammation in AAA formation. APPROACH AND RESULTS: Apoptosis-associated speck-like protein containing a caspase recruitment domain is highly expressed in adventitial macrophages in human and murine AAA tissues. Using an established mouse model of AAA induced by continuous infusion of angiotensin II in Apoe(-/-) mice, NLR family pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein containing a caspase recruitment domain, and caspase-1 deficiency in Apoe(-/-) mice were shown to decrease the incidence, maximal diameter, and severity of AAA along with adventitial fibrosis and inflammatory responses significantly, such as inflammatory cell infiltration and cytokine expression in the vessel wall. NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain, and caspase-1 deficiency in Apoe(-/-) mice also reduced elastic lamina degradation and metalloproteinase activation in the early phase of AAA formation. Furthermore, angiotensin II stimulated generation of mitochondria-derived reactive oxygen species in the adventitial macrophages, and this mitochondria-derived reactive oxygen species generation was inhibited by NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain, and caspase-1 deficiency. In vitro experiments revealed that angiotensin II stimulated the NLRP3 inflammasome activation and subsequent interleukin-1ß release in macrophages, and this activation was mediated through an angiotensin type I receptor/mitochondria-derived reactive oxygen species-dependent pathway. CONCLUSIONS: Our results demonstrate the importance of the NLRP3 inflammasome in the initial inflammatory responses in AAA formation, indicating its potential as a novel therapeutic target for preventing AAA progression.


Asunto(s)
Angiotensina II , Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/metabolismo , Inflamasomas/metabolismo , Activación de Macrófagos , Macrófagos/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo , Anciano , Animales , Aorta Abdominal/inmunología , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/inmunología , Aneurisma de la Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/prevención & control , Apolipoproteínas E , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Adaptadoras de Señalización CARD , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 1/deficiencia , Caspasa 1/genética , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Fibrosis , Humanos , Inflamasomas/genética , Inflamasomas/inmunología , Mediadores de Inflamación/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factores de Tiempo
4.
J Immunol ; 192(9): 4342-51, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24696236

RESUMEN

Inflammation plays a key role in the pathophysiology of hepatic ischemia-reperfusion (I/R) injury. However, the mechanism by which hepatic I/R induces inflammatory responses remains unclear. Recent evidence indicates that a sterile inflammatory response triggered by I/R is mediated through a multiple-protein complex called the inflammasome. Therefore, we investigated the role of the inflammasome in hepatic I/R injury and found that hepatic I/R stimuli upregulated the inflammasome-component molecule, nucleotide-binding oligomerization domain-like receptor (NLR) family pyrin domain-containing 3 (NLRP3), but not apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC). NLRP3(-/-) mice, but not ASC(-/-) and caspase-1(-/-) mice, had significantly less liver injury after hepatic I/R. NLRP3(-/-) mice showed reduced inflammatory responses, reactive oxygen species production, and apoptosis in I/R liver. Notably, infiltration of neutrophils, but not macrophages, was markedly inhibited in the I/R liver of NLRP3(-/-) mice. Bone marrow transplantation experiments showed that NLRP3 not only in bone marrow-derived cells, but also in non-bone marrow-derived cells contributed to liver injury after I/R. In vitro experiments revealed that keratinocyte-derived chemokine-induced activation of heterotrimeric G proteins was markedly diminished. Furthermore, NLRP3(-/-) neutrophils decreased keratinocyte-derived chemokine-induced concentrations of intracellular calcium elevation, Rac activation, and actin assembly formation, thereby resulting in impaired migration activity. Taken together, NLRP3 regulates chemokine-mediated functions and recruitment of neutrophils, and thereby contributes to hepatic I/R injury independently of inflammasomes. These findings identify a novel role of NLRP3 in the pathophysiology of hepatic I/R injury.


Asunto(s)
Proteínas Portadoras/inmunología , Hígado/inmunología , Neutrófilos/inmunología , Daño por Reperfusión/inmunología , Animales , Apoptosis/inmunología , Western Blotting , Proteínas Portadoras/metabolismo , Quimiotaxis de Leucocito , Citometría de Flujo , Inmunohistoquímica , Inflamasomas/inmunología , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Neutrófilos/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Daño por Reperfusión/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Am J Pathol ; 184(5): 1287-98, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24606883

RESUMEN

Inflammation plays a crucial role in the pathophysiological characteristics of chronic kidney disease; however, the inflammatory mechanisms underlying the chronic kidney disease process remain unclear. Recent evidence indicates that sterile inflammation triggered by tissue injury is mediated through a multiprotein complex called the inflammasome. Therefore, we investigated the role of the inflammasome in the development of chronic kidney disease using a murine unilateral ureteral obstruction (UUO) model. Inflammasome-related molecules were up-regulated in the kidney after UUO. Apoptosis-associated speck-like protein containing a caspase recruitment domain deficiency significantly reduced inflammatory responses, such as inflammatory cell infiltration and cytokine expression, and improved subsequent renal injury and fibrosis. Furthermore, apoptosis-associated speck-like protein containing a caspase recruitment domain was specifically up-regulated in collecting duct (CD) epithelial cells of the UUO-treated kidney. In vitro experiments showed that extracellular adenosine triphosphate (ATP) induced inflammasome activation in CD epithelial cells through P2X7-potassium efflux and reactive oxygen species-dependent pathways. These results demonstrate the molecular basis for the inflammatory response in the process of chronic kidney disease and suggest the CD inflammasome as a potential therapeutic target for preventing chronic kidney disease progression.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Inflamación/complicaciones , Túbulos Renales Colectores/patología , Obstrucción Ureteral/complicaciones , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Adaptadoras de Señalización CARD , Citocinas/metabolismo , Fibrosis , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones Endogámicos C57BL , Obstrucción Ureteral/metabolismo , Obstrucción Ureteral/patología
6.
Circulation ; 123(6): 594-604, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21282498

RESUMEN

Background- Inflammation plays a key role in the pathophysiology of myocardial ischemia/reperfusion (I/R) injury; however, the mechanism by which myocardial I/R induces inflammation remains unclear. Recent evidence indicates that a sterile inflammatory response triggered by tissue damage is mediated through a multiple-protein complex called the inflammasome. Therefore, we hypothesized that the inflammasome is an initial sensor for danger signal(s) in myocardial I/R injury. Methods and Results- We demonstrate that inflammasome activation in cardiac fibroblasts, but not in cardiomyocytes, is crucially involved in the initial inflammatory response after myocardial I/R injury. We found that inflammasomes are formed by I/R and that its subsequent activation of inflammasomes leads to interleukin-1ß production, resulting in inflammatory responses such as inflammatory cell infiltration and cytokine expression in the heart. In mice deficient for apoptosis-associated speck-like adaptor protein and caspase-1, these inflammatory responses and subsequent injuries, including infarct development and myocardial fibrosis and dysfunction, were markedly diminished. Bone marrow transplantation experiments with apoptosis-associated speck-like adaptor protein-deficient mice revealed that inflammasome activation in bone marrow cells and myocardial resident cells such as cardiomyocytes or cardiac fibroblasts plays an important role in myocardial I/R injury. In vitro experiments revealed that hypoxia/reoxygenation stimulated inflammasome activation in cardiac fibroblasts, but not in cardiomyocytes, and that hypoxia/reoxygenation-induced activation was mediated through reactive oxygen species production and potassium efflux. Conclusions- Our results demonstrate the molecular basis for the initial inflammatory response after I/R and suggest that the inflammasome is a potential novel therapeutic target for preventing myocardial I/R injury.


Asunto(s)
Fibroblastos/metabolismo , Inflamasomas/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Animales , Caspasa 1/metabolismo , Citocinas/biosíntesis , Humanos , Inflamación/metabolismo , Interleucina-1beta/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Potasio/metabolismo , Especies Reactivas de Oxígeno/metabolismo
7.
Biochem Biophys Res Commun ; 420(1): 72-7, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22402284

RESUMEN

OBJECTIVE: Several reports describe the role of interleukin (IL)-17 in the development of atherosclerosis; however, its precise role remains controversial. We generated double-deficient mice for apolipoprotein E (apoE) and IL-17 (apoE(-/-)IL-17(-/-) mice) and investigated the effect of IL-17 deficiency on vascular inflammation and atherosclerosis. METHODS AND RESULTS: Atherosclerotic plaque areas in apoE(-/-)IL-17(-/-) mice fed a Western diet (WD) were significantly reduced compared with those in apoE(-/-) mice. No significant differences in plasma lipid profiles were observed between apoE(-/-) and apoE(-/-)IL-17(-/-) mice. The number of infiltrated macrophages in the plaques was significantly decreased in WD-fed apoE(-/-)IL-17(-/-) mice compared with WD-fed apoE(-/-) mice, whereas vascular smooth muscle cell content was not altered by IL-17 deficiency. Expression of inflammatory cytokines (MCP-1, IL-1ß, IL-6, IFN-γ, and IL-12 p40) and scavenger receptors (Msr-1, Scarb1, and Olr1) in the plaques was inhibited in WD-fed apoE(-/-)IL-17(-/-) mice. Furthermore, expression of inducible nitric oxide (M1 marker) and arginase-1 (M2 marker) was inhibited in WD-fed apoE(-/-)IL-17(-/-) mice. CONCLUSION: Our results indicate that IL-17 deficiency reduces vascular inflammation and atherosclerosis and that modulation of IL-17 could be a potential target for prevention and treatment of atherosclerosis.


Asunto(s)
Aterosclerosis/inmunología , Interleucina-17/genética , Vasculitis/inmunología , Animales , Apolipoproteínas E/genética , Aterosclerosis/genética , Citocinas/biosíntesis , Dieta/efectos adversos , Modelos Animales de Enfermedad , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Vasculitis/genética
8.
Biochem Biophys Res Commun ; 425(2): 162-8, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22819845

RESUMEN

OBJECTIVE: Recent investigations have suggested that the inflammasome plays a role in the development of vascular inflammation and atherosclerosis; however, its precise role remains controversial. We produced double-deficient mice for apolipoprotien E (Apoe) and caspase-1 (Casp1), a key component molecule of the inflammasome, and investigated the effect of caspase-1 deficiency on vascular inflammation and atherosclerosis. METHODS AND RESULTS: Atherosclerotic plaque areas in whole aortas and aortic root of Western diet (WD)-fed Apoe(-/-)Casp1(-/-) mice were significantly reduced compared to those in Apoe(-/-) mice. The amount of macrophages and vascular smooth muscle cells in the plaques was also reduced in Apoe(-/-)Casp1(-/-) mice. No significant differences in plasma lipid profiles and body weight change were observed between these mice. Expression of interleukin (IL)-1ß in the plaques as well as plasma levels of IL-1ß, IL-1α, IL-6, CCL2, and TNF-α, in Apoe(-/-)Casp1(-/-) mice were lower than those in Apoe(-/-) mice. In vitro experiments showed that calcium phosphate crystals induced caspase-1 activation and secretion of IL-1ß and IL-1α in macrophages. CONCLUSION: Our findings suggest that caspase-1 plays a critical role in vascular inflammation and atherosclerosis, and that modulation of caspase-1 could be a potential target for prevention and treatment of atherosclerosis.


Asunto(s)
Aterosclerosis/enzimología , Caspasa 1/fisiología , Dieta/efectos adversos , Vasculitis/enzimología , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/etiología , Aterosclerosis/genética , Caspasa 1/genética , Inflamasomas/metabolismo , Macrófagos/enzimología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Vasculitis/etiología , Vasculitis/genética
9.
J Reprod Dev ; 58(4): 432-7, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22498815

RESUMEN

Primordial germ cells (PGCs) are embryonic precursors of germline cells with potential applications in genetic conservation, transgenic animal production and germline stem cell research. These lines of research would benefit from improved germline transmission of transplanted PGCs in chimeric chickens. We therefore evaluated the effects of pretransplant X-irradiation of recipient embryos on the efficacy of germline transmission of donor PGCs in chimeric chickens. Intact chicken eggs were exposed to X-ray doses of 3, 6 and 9 Gy (dose rate = 0.12 Gy/min) after 52 h of incubation. There was no significant difference in hatching rate between the 3-Gy-irradiated group and the nonirradiated control group (40.0 vs. 69.6%), but the hatching rate in the 6-Gy-irradiated group (28.6%) was significantly lower than in the control group (P<0.05). No embryos irradiated with 9 Gy of X-rays survived to hatching. X-irradiation significantly reduced the number of endogenous PGCs in the embryonic gonads at stage 27 in a dose-dependent manner compared with nonirradiated controls. The numbers of endogenous PGCs in the 3-, 6- and 9-Gy-irradiated groups were 21.0, 9.6 and 4.6% of the nonirradiated control numbers, respectively. Sets of 100 donor PGCs were subsequently transferred intravascularly into embryos irradiated with 3 Gy X-rays and nonirradiated control embryos. Genetic cross-test analysis revealed that the germline transmission rate in the 3-Gy-irradiated group was significantly higher than in the control group (27.5 vs. 5.6%; P<0.05). In conclusion, X-irradiation reduced the number of endogenous PGCs and increased the germline transmission of transferred PGCs in chimeric chickens.


Asunto(s)
Embrión de Pollo/efectos de la radiación , Desarrollo Embrionario/efectos de la radiación , Células Germinativas/efectos de la radiación , Células Germinativas/trasplante , Mutación de Línea Germinal/efectos de la radiación , Gónadas/efectos de la radiación , Quimera por Radiación/embriología , Crianza de Animales Domésticos/métodos , Animales , Animales Endogámicos , Embrión de Pollo/citología , Embrión de Pollo/embriología , Embrión de Pollo/crecimiento & desarrollo , Pollos , Relación Dosis-Respuesta en la Radiación , Estudios de Factibilidad , Femenino , Células Germinativas/citología , Gónadas/citología , Gónadas/embriología , Supervivencia de Injerto , Inmunohistoquímica/veterinaria , Masculino , Quimera por Radiación/crecimiento & desarrollo , Efectos de la Radiación , Análisis de Supervivencia , Rayos X
11.
Biol Reprod ; 83(1): 130-7, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20357273

RESUMEN

We report a novel technique for almost complete replacement of the recipient germline with donor germ cells in the chicken. Busulfan solubilized in a sustained-release emulsion was injected into the yolk of fertile eggs before incubation. A dose of 100 microg was found to provide the best outcome in terms of reducing the number of endogenous primordial germ cells (PGCs) in embryonic gonads (0.6% of control numbers) and hatchability (36.4%). This was applied for preparing partially sterilized embryos to serve as recipients for the transfer of exogenous PGCs. Immunohistochemical analysis showed that the proportion of donor PGCs in busulfan-treated embryos was significantly higher than in controls (98.6% vs. 6.4%). Genetic cross-test analysis revealed that the germline transmission rate in busulfan-treated chickens was significantly higher than in controls (99.5% vs. 6.0%). Of 11 chimeras, 7 produced only donor-derived progenies, suggesting that these produced only donor-derived gametes in the recipient's gonads. This novel germline replacement technique provides a powerful tool for studying germline differentiation, for generating transgenic individuals, and for conserving genetic resources in birds.


Asunto(s)
Quimerismo , Células Germinativas/trasplante , Quimera por Trasplante , Animales , Busulfano/farmacología , Embrión de Pollo , Pollos , Desarrollo Embrionario/efectos de los fármacos , Femenino , Gónadas/efectos de los fármacos , Gónadas/embriología , Masculino , Agonistas Mieloablativos/farmacología , Esterilización Reproductiva/métodos
12.
Reprod Fertil Dev ; 22(8): 1237-46, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20883649

RESUMEN

The unique accessibility of chicken primordial germ cells (PGCs) during early development provides the opportunity to combine the reproduction of live animals with genetic conservation. Male and female Gifujidori fowl (GJ) PGCs were collected from the blood of early embryos, and cryopreserved in liquid nitrogen for >6 months until transfer. Manipulated GJ embryos were cultured until hatching; fertility tests indicated that they had normal reproductive abilities. Embryos from two lines of White Leghorn (24HS, ST) were used as recipients for chimera production following blood removal. The concentration of PGCs in the early embryonic blood of 24HS was significantly higher than in ST (P < 0.05). Frozen-thawed GJ PGCs were microinjected into the bloodstream of same-sex recipients. Offspring originating from GJ PGCs in ST recipients were obtained with a higher efficiency than those originating from GJ PGCs in 24HS recipients (23.3% v. 3.1%). Additionally, GJ progeny were successfully regenerated by crossing germline chimeras of the ST group. In conclusion, the cryogenic preservation of PGCs from early chicken embryos was combined with the conservation of live animals.


Asunto(s)
Pollos/genética , Criopreservación/veterinaria , Especies en Peligro de Extinción , Células Germinativas/trasplante , Animales , Embrión de Pollo , Quimera , Técnicas de Cultivo de Embriones/veterinaria , Femenino , Fertilidad , Sangre Fetal/citología , Inseminación Artificial/veterinaria , Masculino , Microinyecciones/veterinaria
13.
Cells Tissues Organs ; 189(5): 348-55, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18840994

RESUMEN

Recently, many results have been reported regarding the pluripotency of bone marrow cells (BMCs) with the aim of benefiting regenerative medicine for humans. Particularly, vessel formation by hematopoietic stem cells or vascular endothelial stem cells which were derived from bone marrow has received considerable interest, since the mechanism of vessel formation has been found to be involved in neoangiogenesis of serious diseases such as cancer. Most work on neoangiogenesis and regeneration has involved mammalian experimental systems, however the avian model is useful since the process of neoangiogenesis and regeneration of vessels can be observed with the whole embryo culture system. We have established a novel system using early chick embryos, where a portion of blood vessels are degenerated by UV irradiation, and vessel regeneration is then studied. Incubated embryos were partially covered with aluminum foil, from the embryonic body to the dorsal marginal vein, and irradiated with UV for 1 min. Donor BMCs were obtained from the femurs and tibias of chicks aged 10 days, fluorescently labeled with PKH26 and injected into the anterior vitelline vein of the recipients. In BMC-treated embryos the donor BMCs were observed around the UV-degenerated vessels, and regeneration of blood vessels occurred, in contrast to the untreated embryos. These results indicate that avian BMCs have the ability to participate in vessel regeneration, and the avian model used here may be a useful tool for studies of vessel neoangiogenesis and repair.


Asunto(s)
Vasos Sanguíneos/fisiología , Vasos Sanguíneos/efectos de la radiación , Células de la Médula Ósea/citología , Regeneración/fisiología , Animales , Vasos Sanguíneos/crecimiento & desarrollo , Células Cultivadas , Embrión de Pollo , Pollos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ingeniería de Tejidos/métodos , Rayos Ultravioleta
14.
Reprod Fertil Dev ; 20(8): 900-7, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19007554

RESUMEN

The aim of the present study was to improve the efficiency of endogenous primordial germ cell (PGC) depletion and to increase the ratio of donor PGCs in the gonads of recipient chicken embryos. A sustained-release emulsion was prepared by emulsifying equal amounts of Ca(2+)- and Mg(2+)-free phosphate-buffered saline containing 10% busulfan solubilised in N,N-dimethylformamide and sesame oil, using a filter. Then, 75 microg per 50 microL busulfan sustained-release emulsion was injected into the yolk. To determine the depletion and repopulation of PGCs in the gonads after 6 days incubation, whole-mount immunostaining was performed. The busulfan sustained-release emulsion significantly reduced the number of endogenous PGCs compared with control (P < 0.05). Moreover, the busulfan sustained-release emulsion significantly depleted endogenous PGCs compared with other previously reported busulfan delivery systems (P < 0.05), but with less variation, suggesting that the sustained-release emulsion delivered a consistent amount of busulfan to the developing chicken embryos. The PGC transfer study showed that the proportion of donor PGCs in the gonads of busulfan sustained-release emulsion-treated embryos after 6 days incubation increased 28-fold compared with control. In conclusion, the results demonstrate that exogenous PGCs are capable of migrating and settling in gonads from which endogenous PGCs have been removed using a busulfan sustained-release emulsion.


Asunto(s)
Alquilantes/farmacología , Busulfano/farmacología , Trasplante de Células/métodos , Embrión de Pollo/efectos de los fármacos , Células Germinativas/efectos de los fármacos , Esterilización Reproductiva , Quimera por Trasplante , Alquilantes/administración & dosificación , Animales , Busulfano/administración & dosificación , Movimiento Celular/efectos de los fármacos , Embrión de Pollo/citología , Preparaciones de Acción Retardada , Desarrollo Embrionario/efectos de los fármacos , Emulsiones , Células Germinativas/citología , Inyecciones
15.
Sci Rep ; 6: 26489, 2016 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-27225830

RESUMEN

NLRP3 inflammasomes recognize non-microbial danger signals and induce release of proinflammatory cytokine interleukin (IL)-1ß, leading to sterile inflammation in cardiovascular disease. Because sterile inflammation is involved in doxorubicin (Dox)-induced cardiotoxicity, we investigated the role of NLRP3 inflammasomes in Dox-induced cardiotoxicity. Cardiac dysfunction and injury were induced by low-dose Dox (15 mg/kg) administration in NLRP3-deficient (NLRP3(-/-)) mice but not in wild-type (WT) and IL-1ß(-/-) mice, indicating that NLRP3 deficiency enhanced the susceptibility to Dox-induced cardiotoxicity independent of IL-1ß. Although the hearts of WT and NLRP3(-/-) mice showed no significant difference in inflammatory cell infiltration, macrophages were the predominant inflammatory cells in the hearts, and cardiac IL-10 production was decreased in Dox-treated NLRP3(-/-) mice. Bone marrow transplantation experiments showed that bone marrow-derived cells contributed to the exacerbation of Dox-induced cardiotoxicity in NLRP3(-/-) mice. In vitro experiments revealed that NLRP3 deficiency decreased IL-10 production in macrophages. Furthermore, adeno-associated virus-mediated IL-10 overexpression restored the exacerbation of cardiotoxicity in the NLRP3(-/-) mice. These results demonstrated that NLRP3 regulates macrophage IL-10 production and contributes to the pathophysiology of Dox-induced cardiotoxicity, which is independent of IL-1ß. Our findings identify a novel role of NLRP3 and provided new insights into the mechanisms underlying Dox-induced cardiotoxicity.


Asunto(s)
Doxorrubicina/toxicidad , Lesiones Cardíacas/inmunología , Interleucina-10/metabolismo , Interleucina-1beta/genética , Macrófagos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Animales , Trasplante de Médula Ósea/efectos adversos , Cardiotoxicidad , Células Cultivadas , Modelos Animales de Enfermedad , Lesiones Cardíacas/inducido químicamente , Lesiones Cardíacas/genética , Humanos , Interleucina-1beta/deficiencia , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia
16.
Endocrinology ; 156(11): 4281-92, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26360504

RESUMEN

Preeclampsia is a pregnancy-specific syndrome characterized by elevated blood pressure, proteinuria, and intrauterine growth restriction (IUGR). Although sterile inflammation appears to be involved, its pathogenesis remains unclear. Recent evidence indicates that sterile inflammation is mediated through the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasomes, composed of NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), and caspase-1. Here we investigated the role of the NLRP3 inflammasomes in the pathogenesis of preeclampsia using Nlrp3(-/-) and Asc(-/-) (Nlrp3 and Asc deficient) pregnant mice. During pregnancy in mice, continuous infusion of high-dose angiotensin II (AngII) induced hypertension, proteinuria, and IUGR, whereas infusion of low-dose AngII caused hypertension alone. AngII-induced hypertension was prevented in Nlrp3(-/-) mice but not in Asc(-/-), indicating that NLRP3 contributes to gestational hypertension independently of ASC-mediated inflammasomes. Although NLRP3 deficiency had no effect on IUGR, it restored the IL-6 up-regulation in the placenta and kidney of AngII-infused mice. Furthermore, treatment with hydralazine prevented the development of gestational hypertension but not IUGR or IL-6 expression in the placenta and kidney. These findings demonstrate that NLRP3 contributes to the development of gestational hypertension independently of the inflammasomes and that IUGR and kidney injury can occur independent of blood pressure elevation during pregnancy.


Asunto(s)
Presión Sanguínea/fisiología , Proteínas Portadoras/metabolismo , Retardo del Crecimiento Fetal/metabolismo , Hipertensión/metabolismo , Angiotensina II , Animales , Presión Sanguínea/efectos de los fármacos , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Femenino , Retardo del Crecimiento Fetal/genética , Hipertensión/inducido químicamente , Hipertensión/genética , Hipertensión Inducida en el Embarazo/inducido químicamente , Hipertensión Inducida en el Embarazo/genética , Hipertensión Inducida en el Embarazo/metabolismo , Inflamasomas/metabolismo , Interleucina-6/metabolismo , Riñón/metabolismo , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Placenta/metabolismo , Embarazo
17.
FEBS Open Bio ; 5: 348-56, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25973362

RESUMEN

Increasing evidence indicates that caspase recruitment domain (CARD)-mediated caspase-1 (CASP1) assembly is an essential process for its activation and subsequent interleukin (IL)-1ß release, leading to the initiation of inflammation. Both CARD16 and CARD17 were previously reported as inhibitory homologs of CASP1; however, their molecular function remains unclear. Here, we identified that oligomerization activity allows CARD16 to function as a CASP1 activator. We investigated the molecular characteristics of CARD16 and CARD17 in transiently transfected HeLa cells. Although both CARD16 and CARD17 interacted with CASP1CARD, only CARD16 formed a homo-oligomer. Oligomerized CARD16 formed a filament-like structure with CASP1CARD and a speck with apoptosis-associated speck-like protein containing a CARD. A filament-like structure formed by CARD16 promoted CASP1 filament assembly and IL-1ß release. In contrast, CARD17 did not form a homo-oligomer or filaments and inhibited CASP1-dependent IL-1ß release. Mutated CARD16D27G, mimicking the CARD17 amino acid sequence, formed a homo-oligomer but failed to form a filament-like structure. Consequently, CARD16D27G weakly promoted CASP1 filament assembly and subsequent IL-1ß release. These results suggest that oligomerized CARD16 promotes CARD-mediated molecular assembly and CASP1 activation.

18.
Sci Rep ; 5: 15883, 2015 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-26515636

RESUMEN

Inflammation plays an important role in the development of obesity and metabolic disorders; however, it has not been fully understood how inflammation occurs and is regulated in their pathogenesis. Low-molecular mass protein-7 (LMP7) is a proteolytic subunit of the immunoproteasome that shapes the repertoire of antigenic peptides on major histocompatibility complex class I molecule. In this study, we investigated the role of LMP7 in the development of obesity and metabolic disorders using LMP7-deficient mice. LMP7 deficiency conveyed resistant to obesity, and improved glucose intolerance and insulin sensitivity in mice fed with high-fat diet (HFD). LMP7 deficiency decreased pancreatic lipase expression, increased fecal lipid contents, and inhibited the increase of plasma triglyceride levels upon oral oil administration or HFD feeding. Using bone marrow-transferred chimeric mice, we found that LMP7 in both bone marrow- and non-bone marrow-derived cells contributes to the development of HFD-induced obesity. LMP7 deficiency decreased inflammatory responses such as macrophage infiltration and chemokine expression while it increased serum adiponection levels. These findings demonstrate a novel role for LMP7 and provide new insights into the mechanisms underlying inflammation in the pathophysiology of obesity and metabolic disorders.


Asunto(s)
Enfermedades Metabólicas/patología , Obesidad/patología , Complejo de la Endopetidasa Proteasomal/genética , Adiponectina/sangre , Tejido Adiposo/metabolismo , Animales , Antígenos CD/metabolismo , Trasplante de Médula Ósea , Quimiocinas/metabolismo , Dieta Alta en Grasa , Metabolismo Energético , Resistencia a la Insulina , Lipasa/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Enfermedades Metabólicas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora , Obesidad/metabolismo , Páncreas/enzimología , Complejo de la Endopetidasa Proteasomal/deficiencia , Grasa Subcutánea Abdominal/diagnóstico por imagen , Triglicéridos/sangre , Microtomografía por Rayos X
19.
Nanotoxicology ; 9(5): 554-67, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25211550

RESUMEN

Despite the increasing commercial use of nanoparticles, little is known about their effects on placental inflammation and pregnancy complications. In this study, nanosilica (NS) particles upregulated the inflammasome component nucleotide-binding oligomerization domain-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) and induced placental inflammation and reactive oxygen species (ROS) generation, resulting in pregnancy complications. Furthermore, NS-induced pregnancy complications were markedly improved in Nlrp3(-/-) mice but not in component apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC)-deficient (Asc(-/-)) mice, indicating the independence of NLRP3 inflammasomes. Pregnancy complications in Nlrp3(-/-) and Asc(-/-) mice phenotypes were dependent on the balance between interleukin (IL)-1α and IL-10. NS-induced pregnancy complications were completely prevented by either inhibition of ROS generation or forced expression of IL-10. Our findings provide important information about NS-induced placental inflammation and pregnancy complications and the novel pathophysiological roles of NLRP3 and ASC in pregnancy.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Portadoras/metabolismo , Inflamasomas/inmunología , Nanopartículas/toxicidad , Placenta/efectos de los fármacos , Complicaciones del Embarazo/inducido químicamente , Dióxido de Silicio/toxicidad , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Adaptadoras de Señalización CARD , Proteínas Portadoras/genética , Femenino , Inflamasomas/metabolismo , Interleucina-10/inmunología , Interleucina-1alfa/inmunología , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Nanopartículas/química , Placenta/inmunología , Placenta/patología , Embarazo , Complicaciones del Embarazo/inmunología , Complicaciones del Embarazo/patología , Especies Reactivas de Oxígeno/metabolismo , Dióxido de Silicio/química
20.
Sci Rep ; 5: 10901, 2015 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-26045078

RESUMEN

Rhabdomyolysis is one of the main causes of community-acquired acute kidney injury (AKI). Although inflammation is involved in the pathogenesis of rhabdomyolysis-induced AKI (RIAKI), little is known about the mechanism that triggers inflammation during RIAKI. Recent evidence has indicated that sterile inflammation triggered by tissue injury can be mediated through multiprotein complexes called the inflammasomes. Therefore, we investigated the role of NLRP3 inflammasomes in the pathogenesis of RIAKI using a glycerol-induced murine rhabdomyolysis model. Inflammasome-related molecules were upregulated in the kidney of RIAKI. Renal tubular injury and dysfunction preceded leukocyte infiltration into the kidney during the early phase of RIAKI, and they were markedly attenuated in mice deficient in NLRP3, ASC, caspase-1, and interleukin (IL)-1ß compared with those in wild-type mice. No difference in leukocyte infiltration was observed between wild-type and NLRP3-deficient mice. Furthermore, NLRP3 deficiency strikingly suppressed the expression of renal injury markers and inflammatory cytokines and apoptosis of renal tubular cells. These results demonstrated that NLRP3 inflammasomes contribute to inflammation, apoptosis, and tissue injury during the early phase of RIAKI and provide new insights into the mechanism underlying the pathogenesis of RIAKI.


Asunto(s)
Lesión Renal Aguda/etiología , Lesión Renal Aguda/metabolismo , Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Rabdomiólisis/complicaciones , Lesión Renal Aguda/patología , Animales , Apoptosis , Biomarcadores , Proteínas Portadoras/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Mediadores de Inflamación/metabolismo , Túbulos Renales/metabolismo , Leucocitos/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA