Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Circ Res ; 135(6): e133-e149, 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-39082135

RESUMEN

BACKGROUND: Prostaglandin I2 synthesized by endothelial COX (cyclooxygenase) evokes potent vasodilation in some blood vessels but is paradoxically responsible for endothelium-dependent constriction (EDC) in others. Prostaglandin I2 production and EDC may be enhanced in diseases such as hypertension. However, how PGIS (prostaglandin I2 synthase) deficiency affects EDC and how this is implicated in the consequent cardiovascular pathologies remain largely unknown. METHODS: Experiments were performed with wild-type, Pgis knockout (Pgis-/-) and Pgis/thromboxane-prostanoid receptor gene (Tp) double knockout (Pgis-/-Tp-/-) mice and Pgis-/- mice transplanted with unfractionated wild-type or Cox-1-/- bone marrow cells, as well as human umbilical arteries. COX-derived prostanoids were measured by high-performance liquid chromatography-mass spectrometry. Vasomotor responses of distinct types of arteries were assessed by isometric force measurement. Parameters of hypertension, vascular remodeling, and cardiac hypertrophy in mice at different ages were monitored. RESULTS: PGF2α, PGE2, and a trace amount of PGD2, but not thromboxane A2 (TxA2), were produced in response to acetylcholine in Pgis-/- or PGIS-inhibited arteries. PGIS deficiency resulted in exacerbation or occurrence of EDC ex vivo and in vivo. Endothelium-dependent hyperpolarization was unchanged, but phosphorylation levels of eNOS (endothelial nitric oxide synthase) at Ser1177 and Thr495 were altered and NO production and the NO-dependent relaxation evoked by acetylcholine were remarkably reduced in Pgis-/- aortas. Pgis-/- mice developed high blood pressure and vascular remodeling at 16 to 17 weeks and subsequently cardiac hypertrophy at 24 to 26 weeks. Meanwhile, blood pressure and cardiac parameters remained normal at 8 to 10 weeks. Additional ablation of TP (TxA2 receptor) not only restrained EDC and the downregulation of NO signaling in Pgis-/- mice but also ameliorated the cardiovascular abnormalities. Stimulation of Pgis-/- vessels with acetylcholine in the presence of platelets led to increased TxA2 generation. COX-1 disruption in bone marrow-derived cells failed to affect the development of high blood pressure and vascular remodeling in Pgis-/- mice though it largely suppressed the increase of plasma TxB2 (TxA2 metabolite) level. CONCLUSIONS: Our study demonstrates that the non-TxA2 prostanoids/TP axis plays an essential role in mediating the augmentation of EDC and cardiovascular disorders when PGIS is deficient, suggesting TP as a promising therapeutic target in diseases associated with PGIS insufficiency.


Asunto(s)
Endotelio Vascular , Oxidorreductasas Intramoleculares , Ratones Endogámicos C57BL , Ratones Noqueados , Prostaglandinas , Vasoconstricción , Animales , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/metabolismo , Ratones , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Prostaglandinas/metabolismo , Humanos , Sistema Enzimático del Citocromo P-450/metabolismo , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/deficiencia , Tromboxano A2/metabolismo , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Enfermedades Cardiovasculares/etiología , Masculino , Receptores de Tromboxanos/metabolismo , Receptores de Tromboxanos/genética , Vasodilatación , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Remodelación Vascular , Transducción de Señal , Ciclooxigenasa 1/deficiencia , Ciclooxigenasa 1/genética , Ciclooxigenasa 1/metabolismo
2.
Oxid Med Cell Longev ; 2021: 6192498, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34306312

RESUMEN

Established studies proved that mechanical compression loading had multiple effects on the biological behavior of the intervertebral disc (IVD). However, the regulating mechanism involved in this process remains unclear. The current study is aimed at exploring the potential bioregulators and signaling pathways involved in the compression-associated biological changes of nucleus pulposus (NP) cells. Tandem mass tag- (TMT-) based quantitative proteomics was exerted to analyze the differentially expressed proteins (DEPs) and signal pathways among the different groups of NP cells cultured under noncompression, low-compression (LC), and high-compression (HC) loading. Eight potential protective bioregulators for the NP cell survival under different compression loading were predicted by the proteomics, among which macrophage migration inhibitory factor (MIF) and oxidative stress-related pathways were selected for further evaluation, due to its similar function in regulating the fate of the cartilage endplate- (CEP-) derived cells. We found that deficiency of MIF accentuates the accumulation of ROS, mitochondrial dysfunction, and senescence of NP cells under overloaded mechanical compression. The potential molecular mechanism involved in this process is related to the mitophagy regulating role of MIF. Our findings provide a better understanding of the regulatory role of mechanical compression on the cellular fate commitment and matrix metabolism of NP, and the potential strategies for treating disc degenerative diseases via using MIF-regulating agents.


Asunto(s)
Oxidorreductasas Intramoleculares/deficiencia , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Núcleo Pulposo/metabolismo , Núcleo Pulposo/patología , Estrés Oxidativo/fisiología , Estrés Mecánico , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Humanos , Degeneración del Disco Intervertebral/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Mitofagia/fisiología , Transducción de Señal/efectos de los fármacos
3.
Nat Commun ; 12(1): 2954, 2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-34012010

RESUMEN

How cancer cells cope with high levels of replication stress during rapid proliferation is currently unclear. Here, we show that macrophage migration inhibitory factor (MIF) is a 3' flap nuclease that translocates to the nucleus in S phase. Poly(ADP-ribose) polymerase 1 co-localizes with MIF to the DNA replication fork, where MIF nuclease activity is required to resolve replication stress and facilitates tumor growth. MIF loss in cancer cells leads to mutation frequency increases, cell cycle delays and DNA synthesis and cell growth inhibition, which can be rescued by restoring MIF, but not nuclease-deficient MIF mutant. MIF is significantly upregulated in breast tumors and correlates with poor overall survival in patients. We propose that MIF is a unique 3' nuclease, excises flaps at the immediate 3' end during DNA synthesis and favors cancer cells evading replication stress-induced threat for their growth.


Asunto(s)
Neoplasias de la Mama/metabolismo , Replicación del ADN/fisiología , Endonucleasas de ADN Solapado/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , ADN/química , ADN/metabolismo , Daño del ADN , ADN Polimerasa III/genética , ADN Polimerasa III/metabolismo , Replicación del ADN/genética , Femenino , Endonucleasas de ADN Solapado/deficiencia , Endonucleasas de ADN Solapado/genética , Técnicas de Inactivación de Genes , Inestabilidad Genómica , Células HCT116 , Humanos , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Factores Inhibidores de la Migración de Macrófagos/genética , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Conformación de Ácido Nucleico , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Fase S , Especificidad por Sustrato
4.
Hum Genet ; 140(8): 1157-1168, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33959807

RESUMEN

Infantile nystagmus syndrome (INS) denominates early-onset, involuntary oscillatory eye movements with different etiologies. Nystagmus is also one of the symptoms in oculocutaneus albinism (OCA), a heterogeneous disease mainly caused by defects in melanin synthesis or melanosome biogenesis. Dopachrome tautomerase (DCT, also called TYRP2) together with tyrosinase (TYR) and tyrosin-related protein 1 (TYRP1) is one of the key enzymes in melanin synthesis. Although DCT´s role in pigmentation has been proven in different species, until now only mutations in TYR and TYRP1 have been found in patients with OCA. Detailed ophthalmological and orthoptic investigations identified a consanguineous family with two individuals with isolated infantile nystagmus and one family member with subtle signs of albinism. By whole-exome sequencing and segregation analysis, we identified the missense mutation c.176G > T (p.Gly59Val) in DCT in a homozygous state in all three affected family members. We show that this mutation results in incomplete protein maturation and targeting in vitro compatible with a partial or total loss of function. Subsequent screening of a cohort of patients with OCA (n = 85) and INS (n = 25) revealed two heterozygous truncating mutations, namely c.876C > A (p.Tyr292*) and c.1407G > A (p.Trp469*), in an independent patient with OCA. Taken together, our data suggest that mutations in DCT can cause a phenotypic spectrum ranging from isolated infantile nystagmus to oculocutaneous albinism.


Asunto(s)
Albinismo Oculocutáneo/genética , Oxidorreductasas Intramoleculares/genética , Melaninas/biosíntesis , Mutación Missense , Nistagmo Congénito/genética , Adolescente , Albinismo Oculocutáneo/diagnóstico , Albinismo Oculocutáneo/enzimología , Albinismo Oculocutáneo/patología , Secuencia de Bases , Calnexina/genética , Calnexina/metabolismo , Niño , Estudios de Cohortes , Consanguinidad , Femenino , Regulación de la Expresión Génica , Células HEK293 , Homocigoto , Humanos , Oxidorreductasas Intramoleculares/deficiencia , Masculino , Melaninas/genética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Monofenol Monooxigenasa/genética , Monofenol Monooxigenasa/metabolismo , Nistagmo Congénito/diagnóstico , Nistagmo Congénito/enzimología , Nistagmo Congénito/patología , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Linaje , Secuenciación del Exoma , Adulto Joven
5.
Biochem Biophys Res Commun ; 546: 124-129, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33582554

RESUMEN

Microsomal prostaglandin (PG) E synthase-1 (mPGES-1) and prostacyclin (PGI2) synthase (PGIS) are PG terminal synthases that work downstream of cyclooxygenase and synthesize PGE2 and PGI2, respectively. Although the involvement of PG receptors in acquired cutaneous immune responses was recently shown, the roles of these PG terminal synthases remain unclear. To identify the pathophysiological roles of mPGES-1 and PGIS in cutaneous immune systems, we applied contact hypersensitivity (CHS) to mPGES-1 and PGIS knockout (KO) mice as a model of acquired immune responses. Mice were treated with 1-fluoro-2,4-dinitrobenzene (DNFB) and evaluated for ear thickness and histopathological features. The results showed that the severity of ear swelling in both gene-deficient mice was much lower than that in wild-type (WT) mice. Histological examination of DNFB-treated ears showed that inflammatory cell infiltration and edema in the dermis were also less apparent in both genotypic mice. LC-MS analysis further showed that the increment in PGE2 levels in DNFB-treated ear tissue was reduced in mPGES-1 KO mice, and that 6-keto PGF1α (a stable metabolite of PGI2) was not detected in PGIS KO mice. Furthermore, we made bone marrow (BM) chimera and found that transplantation of WT mouse-derived BM cells restored the impaired CHS response in mPGES-1 KO mice but did not restore the response in PGIS KO mice. These results indicated that mPGES-1 in BM-derived cells and PGIS in non-BM-derived cells might play critical roles in DNFB-induced CHS. mPGES-1-derived PGE2 and PGIS-derived PGI2 might coordinately promote acquired cutaneous immune responses.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Dermatitis por Contacto/enzimología , Oxidorreductasas Intramoleculares/metabolismo , Prostaglandina-E Sintasas/metabolismo , Traslado Adoptivo , Animales , Células de la Médula Ósea , Sistema Enzimático del Citocromo P-450/deficiencia , Sistema Enzimático del Citocromo P-450/genética , Dermatitis por Contacto/etiología , Dermatitis por Contacto/genética , Dinitrofluorobenceno/efectos adversos , Oído/patología , Femenino , Interferón gamma/metabolismo , Interleucinas/metabolismo , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/genética , Ratones , Ratones Noqueados , Prostaglandina-E Sintasas/deficiencia , Prostaglandina-E Sintasas/genética , Prostaglandinas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-22
6.
Pharmacol Res ; 157: 104828, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32339783

RESUMEN

Second hand smoke exposure increases the prevalence of chronic diseases partly attributed to inflammatory responses. Macrophage migration inhibitory factor (MIF), a proinflammatory cytokine, is involved in the pathogenesis of multiple diseases although its role in second hand smoke exposure-induced cardiac anomalies remains elusive. This study evaluated the impact of MIF knockout on side-stream smoke exposure-induced cardiac pathology and underlying mechanisms. Adult WT and MIF knockout (MIFKO) mice were placed in a chamber exposed to cigarette smoke for 1 h daily for 60 consecutive days. Echocardiographic, cardiomyocyte function and intracellular Ca2+ handling were evaluated. Autophagy, mitophagy and apoptosis were examined using western blot. DHE staining was used to evaluate superoxide anion (O2-) generation. Masson trichrome staining was employed to assess interstitial fibrosis. Our data revealed that MIF knockout accentuated side-stream smoke-induced cardiac anomalies in fractional shortening, cardiomyocyte function, intracellular Ca2+ homeostasis, myocardial ultrastructure and mitochondrial content along with overt apoptosis and O2- generation. In addition, unfavorable effects of side-stream smoke were accompanied by excessive formation of autophagolysosome and elevated TFEB, the effect of which was exacerbated by MIF knockout. Recombinant MIF rescued smoke extract-induced myopathic anomalies through promoting AMPK activation, mitophagy and lysosomal function. Taken together, our data suggest that MIF serves as a protective factor against side-stream smoke exposure-induced myopathic changes through facilitating mitophagy and autophagolysosome formation.


Asunto(s)
Oxidorreductasas Intramoleculares/deficiencia , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Mitocondrias Cardíacas/metabolismo , Mitofagia , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Contaminación por Humo de Tabaco , Disfunción Ventricular Izquierda/metabolismo , Función Ventricular Izquierda , Animales , Señalización del Calcio , Modelos Animales de Enfermedad , Fibrosis , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Cardíacas/ultraestructura , Miocitos Cardíacos/ultraestructura , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología
7.
Prostaglandins Other Lipid Mediat ; 149: 106429, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32145387

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is an emerging risk factor for type 2 diabetes mellitus, cardiovascular disease, and all-cause mortality. Previously, we demonstrated that lipocalin-type prostaglandin D2 synthase (L-PGDS) knockout mice show increased glucose intolerance and accelerated atherosclerosis. In the present study, we investigated the role of L-PGDS in mediating NAFLD utilizing L-PGDS knockout (KO) and control C57BL/6 mice fed either low fat (LFD) or high fat diet (HFD) for 14 weeks. Our present study demonstrates that L-PGDS KO mice remain slightly lighter in weight compared to control mice, yet develop NAFLD faster and eventually progress to the more severe non-alcoholic steatohepatitis (NASH). We found increased lipid accumulation in the liver of KO mice over time on both diets, as compared to control mice. The L-PGDS KO mice showed elevated fasting glucose and insulin levels and developed insulin resistance on both LFD and HFD. Lipogenesis marker proteins such as SREBP-1c and LXRα were increased in L-PGDS KO mice after 14 weeks on both diets, when compared to control mice. We replicated our in vivo findings in vitro using HepG2 cells treated with a combination of free fatty acids (oleic and palmitic acid) and exposure to a L-PGDS inhibitor and prostaglandin D2 receptor (DP1) antagonists. We conclude that the absence or inhibition of L-PGDS results in dyslipidemia, altered expression of lipogenesis genes and the acceleration of NAFLD to NASH, independent of diet and obesity. We propose L-PGDS KO mice as a useful model to explore the pathogenesis of NAFLD and NASH, and L-PGDS as a potential therapeutic target for treatment.


Asunto(s)
Dislipidemias/genética , Eliminación de Gen , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/genética , Lipocalinas/genética , Enfermedad del Hígado Graso no Alcohólico/genética , Animales , Progresión de la Enfermedad , Dislipidemias/enzimología , Dislipidemias/metabolismo , Metabolismo Energético/genética , Regulación de la Expresión Génica/genética , Glucosa/metabolismo , Células Hep G2 , Homeostasis/genética , Humanos , Metabolismo de los Lípidos/genética , Lipogénesis/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/enzimología , Enfermedad del Hígado Graso no Alcohólico/metabolismo
8.
Biomed Pharmacother ; 125: 109919, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32062385

RESUMEN

BACKGROUND: Macrophage migration inhibitory factor (MIF) is an important pro-inflammatory cytokine implicated in sepsis, rheumatoid arthritis and other diseases. However, the role of MIF in acute pancreatitis (AP) remains unclear. This study aims to explore the role of MIF in the pathogenesis of AP using MIF-/- mice (referred to as KO) and the biological effects of pharmacological inhibition of MIF in l-arginine induced AP. METHODS: AP was induced in C57BL/6 wild-type (referred to as WT) and KO mice by administration of l-arginine. The severity of AP was assessed by serum analysis of amylase and lipase, and of these pro-inflammatory cytokines TNF-α and IL-1ß. Histological hematoxylin and eosin (H&E) and immunohistochemical staining of pancreatic tissues were examined for inflammation and expression of pro-inflammatory mediators. We also investigated the biological effects of pharmacological inhibition of MIF activity using ISO-1((S,R)-3-(4-hydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid methyl ester). RESULTS: At 72 h after the induction of AP with l-arginine, significantly lower levels of serum amylase, lipase, TNF-α, and IL-1ß were observed in KO mice when compared with WT controls. Histological examination further showed protective effects against pancreatic tissue damage and inflammation, with pancreatic expression of TNF-α, IL-1ß and NF-κB p65 markedly reduced. Pharmacological inhibition of MIF activity with ISO-1 markedly mirrored the protective effect seen in the KO AP model providing further evidence that MIF is involved in the pathogenesis of AP. CONCLUSION: Our data provided strong evidence for the participation of MIF in the pathogenesis of AP and subsequent inflammatory response. The genetic ablation of MIF or its inhibition with pharmacological agents significantly ameliorated the severity of AP.


Asunto(s)
Modelos Animales de Enfermedad , Eliminación de Gen , Oxidorreductasas Intramoleculares/deficiencia , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Pancreatitis/metabolismo , Pancreatitis/prevención & control , Índice de Severidad de la Enfermedad , Animales , Femenino , Inflamación/genética , Inflamación/metabolismo , Inflamación/prevención & control , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pancreatitis/genética
9.
Methods Mol Biol ; 2080: 57-66, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31745871

RESUMEN

Phenotyping cells by flow cytometry is a powerful way to identify cell type and any morphological changes during cell culture. The staining procedure used in this chapter enables the characterization of mouse macrophages by a flow cytometry antibody panel which can be used for both bone marrow-derived macrophages (BMM) and macrophages derived from other tissues, such as the mouse spleen or peritoneal cavity. The surface and intracellular staining methods are versatile and can be applied to flow cytometry staining of several different cell types by changing the surface markers used with knowledge of which receptors are expressed on different cell types.


Asunto(s)
Citometría de Flujo , Inmunofenotipificación , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Animales , Biomarcadores , Oxidorreductasas Intramoleculares/deficiencia , Activación de Macrófagos/inmunología , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Ratones , Ratones Noqueados , Fenotipo
10.
Mech Ageing Dev ; 182: 111126, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31381889

RESUMEN

The age-induced, exponential accumulation of mitochondrial DNA (mtDNA) deletion mutations contributes to muscle fiber loss. The causes of these mutations are not known. Systemic inflammation is associated with decreased muscle mass in older adults and is implicated in the formation of sporadic mtDNA deletions. Macrophage migration inhibitory factor knockout (MIF-KO) mice are long-lived with decreased inflammation. We hypothesized that aged MIF-KO mice would have lower mtDNA deletion frequencies and fewer electron transport chain (ETC) deficient fibers. We measured mtDNA copy number and mutation frequency as well as the number and length of ETC deficient fibers in 22-month old MIF-KO and F2 hybrid control mice. We also measured mtDNA copy number and deletion frequency in female UM-HET3 mice, a strain whose lifespan matches the MIF-KO mice. We did not observe a significant effect of MIF ablation on muscle mtDNA deletion frequency. There was a significantly lower mtDNA copy number in the MIF-KO mice and the lifespan-matched UM-HET3 mice compared to the F2 hybrids, suggesting the importance of genetic background in mtDNA copy number control. Our data do not support a definitive role for MIF in age-induced mtDNA deletions.


Asunto(s)
Senescencia Celular , Variaciones en el Número de Copia de ADN , ADN Mitocondrial/metabolismo , Oxidorreductasas Intramoleculares/deficiencia , Longevidad , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Macrófagos/metabolismo , Animales , ADN Mitocondrial/genética , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Ratones , Ratones Noqueados
11.
Clin Sci (Lond) ; 133(5): 665-680, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30804219

RESUMEN

Ischemic preconditioning (IPC) is an endogenous protection strategy against myocardial ischemia-reperfusion (I/R) injury. Macrophage migration inhibitory factor (MIF) released from the myocardium subjected to brief periods of ischemia confers cardioprotection. We hypothesized that MIF plays an essential role in IPC-induced cardioprotection. I/R was induced either ex vivo or in vivo in male wild-type (WT) and MIF knockout (MIFKO) mice with or without proceeding IPC (three cycles of 5-min ischemia and 5-min reperfusion). Indices of myocardial injury, regional inflammation and cardiac function were determined to evaluate the extent of I/R injury. Activations of the reperfusion injury salvage kinase (RISK) pathway, AMP-activated protein kinase (AMPK) and their downstream components were investigated to explore the underlying mechanisms. IPC conferred prominent protection in WT hearts evidenced by reduced infarct size (by 33-35%), myocyte apoptosis and enzymatic markers of tissue injury, ROS production, inflammatory cell infiltration and MCP1/CCR2 expression (all P<0.05). IPC also ameliorated cardiac dysfunction both ex vivo and in vivo These protective effects were abolished in MIFKO hearts. Notably, IPC mediated further activations of RISK pathway, AMPK and the membrane translocation of GLUT4 in WT hearts. Deletion of MIF blunted these changes in response to IPC, which is the likely basis for the absence of protective effects of IPC against I/R injury. In conclusion, MIF plays a critical role in IPC-mediated cardioprotection under ischemic stress by activating RISK signaling pathway and AMPK. These results provide an insight for developing a novel therapeutic strategy that target MIF to protect ischemic hearts.


Asunto(s)
Oxidorreductasas Intramoleculares/metabolismo , Precondicionamiento Isquémico Miocárdico/métodos , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Apoptosis , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Factores Inhibidores de la Migración de Macrófagos/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Receptores CCR2/metabolismo , Transducción de Señal , Remodelación Ventricular
12.
Microb Cell Fact ; 17(1): 12, 2018 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-29374483

RESUMEN

BACKGROUND: We previously developed an E. coli strain that overproduces medium-chain methyl ketones for potential use as diesel fuel blending agents or as flavors and fragrances. To date, the strain's performance has been optimized during growth with glucose. However, lignocellulosic biomass hydrolysates also contain a substantial portion of hemicellulose-derived xylose, which is typically the second most abundant sugar after glucose. Commercialization of the methyl ketone-producing technology would benefit from the increased efficiency resulting from simultaneous, rather than the native sequential (diauxic), utilization of glucose and xylose. RESULTS: In this study, genetic manipulations were performed to alleviate carbon catabolite repression in our most efficient methyl ketone-producing strain. A strain engineered for constitutive expression of xylF and xylA (involved in xylose transport and metabolism) showed synchronized glucose and xylose consumption rates. However, this newly acquired capability came at the expense of methyl ketone titer, which decreased fivefold. Further efforts were made to improve methyl ketone production in this strain, and we found that two strategies were effective at enhancing methyl ketone titer: (1) chromosomal deletion of pgi (glucose-6-phosphate isomerase) to increase intracellular NADPH supply and (2) downregulation of CRP (cAMP receptor protein) expression by replacement of the native RBS with an RBS chosen based upon mutant library screening results. Combining these strategies resulted in the most favorable overall phenotypes for simultaneous glucose-xylose consumption without compromising methyl ketone titer at both 1 and 2% total sugar concentrations in shake flasks. CONCLUSIONS: This work demonstrated a strategy for engineering simultaneous utilization of C6 and C5 sugars in E. coli without sacrificing production of fatty acid-derived compounds.


Asunto(s)
Disacáridos/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Cetonas/metabolismo , Técnicas de Cultivo Celular por Lotes , Represión Catabólica , Sistema Enzimático del Citocromo P-450/deficiencia , Sistema Enzimático del Citocromo P-450/genética , Proteínas de Escherichia coli/genética , Fermentación , Glucosa/metabolismo , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/genética , Cetonas/análisis , Ingeniería Metabólica/métodos , Proteínas/genética , Xilosa/metabolismo
13.
J Cell Physiol ; 233(2): 1490-1499, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28574571

RESUMEN

To obtain stable outcomes in regenerative medicine, understanding and controlling immunological responses in transplanted tissues are of great importance. In our previous study, auricular chondrocytes in tissue-engineered cartilage transplanted in mice were shown to express immunological factors, including macrophage migration inhibitory factor (MIF). Since MIF exerts pleiotropic functions, in this study, we examined the roles of MIF in cartilage regenerative medicine. We made tissue-engineered cartilage consisting of auricular chondrocytes of C57BL/6J mouse, atellocollagen gel and a PLLA scaffold, and transplanted the construct subcutaneously in a syngeneic manner. Localization of MIF was prominent in cartilage areas of tissue-engineered cartilage at 2 weeks after transplantation, though it became less apparent by 8 weeks. Co-culture with RAW264 significantly increased the expression of MIF in chondrocytes, suggesting that the transplanted chondrocytes in tissue-engineered cartilage could enhance the expression of MIF by stimulation of surrounding macrophages. When MIF was added in the culture of chondrocytes, the expression of type II collagen was increased, indicating that MIF could promote the maturation of chondrocytes. Meanwhile, toluidine blue staining of constructs containing wild type (Mif+/+) chondrocytes showed increased metachromasia compared to MIF-knockout (Mif-/-) constructs at 2 weeks. However, this tendency was reversed by 8 weeks, suggesting that the initial increase in cartilage maturation in Mif+/+ constructs deteriorated by 8 weeks. Since the Mif+/+ constructs included more iNOS-positive inflammatory macrophages at 2 weeks, MIF might induce an M1 macrophage-polarized environment, which may eventually worsen the maturation of tissue-engineered cartilage in the long term.


Asunto(s)
Comunicación Celular , Condrocitos/metabolismo , Cartílago Auricular/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Macrófagos/metabolismo , Ingeniería de Tejidos/métodos , Animales , Proliferación Celular , Condrocitos/trasplante , Condrogénesis , Técnicas de Cocultivo , Colágeno/metabolismo , Cartílago Auricular/citología , Cartílago Auricular/trasplante , Geles , Humanos , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Factores Inhibidores de la Migración de Macrófagos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fenotipo , Poliésteres/química , Células RAW 264.7 , Transducción de Señal , Factores de Tiempo , Andamios del Tejido
14.
Cytokine ; 99: 214-224, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28780379

RESUMEN

BACKGROUND: Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine that plays an important role in the pathogenesis of type 2 diabetes mellitus (T2DM). Although the effect of high glucose on liver function has been described, the role of MIF in hepatic mitochondrial function during T2DM has not been studied. OBJECTIVE: We examine the influence of MIF to hepatic mitochondrial function in T2DM mouse model. METHODS: WT and Mif-/- BALB/c mice were treated with a single dose of streptozotocin (STZ). After an 8-week follow-up, serum glucose, proinflammatory cytokines, C-reactive protein (CRP), alanine aminotransferase (ALT) and aspartate aminotransferase (AST) enzyme quantification, and liver histological analyses were performed. Liver mitochondria were extracted, and mitochondrial function was evaluated by oximetry, swelling and peroxide production. RESULTS: Following treatment with STZ, WT mice (WT/STZ) developed significant hyperglycemia and high serum levels of MIF, tumor necrosis factor (TNF)-α, interleukin-ß (IL-ß), and CRP. Liver damage enzymes ALT and AST were found at high levels. In contrast, Mif-/-STZ lacked serum MIF levels and showed smaller increases in blood glucose, less TNF-α, IL-1ß, CPR, ALT and AST, and failure to develop clinical signs of disease compared to the WT/STZ group. Mitochondria extracted from the Mif-/-STZ liver showed similar respiratory control (RC) to WT/STZ or healthy mice with glutamate/malate or succinate as substrates. The four respiratory chain complexes also had comparable activities. WT/STZ-isolated mitochondria showed low swelling with calcium compared to mitochondria from Mif-/-STZ or healthy mice. Peroxide production was comparable in all groups. CONCLUSION: These results show although high systemic levels of MIF contribute to the development of T2DM pathology, the liver mitochondria remain unaltered. Importantly, the absence of MIF reduced the pathology of T2DM, also without altering liver mitochondrial function. These support MIF as a therapeutic target for the treatment of this disease in humans.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Mediadores de Inflamación/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Hígado/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Mitocondrias/metabolismo , Animales , Proteína C-Reactiva/metabolismo , Respiración de la Célula , Citocromos/metabolismo , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/patología , Electrodos , Hiperglucemia/complicaciones , Hiperglucemia/patología , Interleucina-1beta/sangre , Oxidorreductasas Intramoleculares/deficiencia , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Hígado/patología , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Masculino , Ratones Endogámicos BALB C , Dilatación Mitocondrial , Oxígeno/metabolismo , Peróxidos/metabolismo , Estreptozocina , Transaminasas/metabolismo , Factor de Necrosis Tumoral alfa/sangre
15.
Prostaglandins Other Lipid Mediat ; 133: 49-52, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28506876

RESUMEN

Prostacyclin (PGI2) synthase (PGIS) and microsomal prostaglandin (PG) E synthase-1 (PGES-1) functionally couple with inducible cyclooxygenase-2 (COX-2) as their upstream enzymes to produce PGI2 and PGE2, respectively. Non-steroidal anti-inflammatory drugs exert their pharmacological effects including antitumor effects by the inhibition of COX-2 and thereby suppress this PG biosynthesis. PGIS is abundantly expressed in vascular endothelial and smooth muscle cells and was shown to be critical for the regulation of platelet aggregation and vascular tone. In addition to its role in vascular regulation, PGIS was shown to be frequently down-regulated in several types of cancers, and the involvement of PGIS in carcinogenesis has been suggested. In this review, we summarize the current understanding of the roles of PGIS and PGIS-derived PGI2 in carcinogenesis.


Asunto(s)
Carcinogénesis , Sistema Enzimático del Citocromo P-450/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Neoplasias/enzimología , Neoplasias/patología , Animales , Sistema Enzimático del Citocromo P-450/deficiencia , Sistema Enzimático del Citocromo P-450/genética , Eliminación de Gen , Regulación Enzimológica de la Expresión Génica , Humanos , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/genética , Neoplasias/genética
16.
Am J Physiol Renal Physiol ; 313(3): F767-F780, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28539339

RESUMEN

Macrophage migration inhibitory factor (MIF) is a cytokine with pleiotropic actions that is produced by several organs and cell types. Depending on the target cell and the inflammatory context, MIF can engage its two component receptor complex CD74 and CD44 and the chemokine receptors CXCR2/4. MIF is constitutively expressed in renal proximal tubular cells, stored in intracellular preformed pools, and released at a low rate. Recently, a second MIF-like protein (i.e., MIF-2/D-DT) has been characterized in mammals. Our study was aimed at examining the role of MIF-2/D-DT, which mediates tissue protection in the heart, in tubular cell regeneration from ischemia-reperfusion injury. We found that Mif-/-, Mif-2-/-, and Cd74-/- mice had significantly worse tubular injury compared with wild-type (WT) control mice and that treatment with MIF-2/D-DT significantly improved recovery of injured epithelial cells. RNAseq analysis of kidney tissue from the ischemia-reperfusion injury model revealed that MIF-2/D-DT treatment stimulates secretory leukocyte proteinase inhibitor (SLPI) and cyclin D1 expression. MIF-2/D-DT additionally activates of eukaryotic initiation factor (eIF) 2α and activating transcription factor (ATF) 4, two transcription factors involved in the integrated stress response (ISR), which is a cellular stress response activated by hypoxia, nutrient deprivation, and oxygen radicals. MIF-2/D-DT also inhibited apoptosis and induced autophagy in hypoxia-treated mouse proximal tubular (MPT) cells. These results indicate that MIF-2/D-DT is an important factor in tubular cell regeneration and may be of therapeutic utility as a regenerative agent in the clinical setting of ischemic acute kidney injury.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Lesión Renal Aguda/metabolismo , Proliferación Celular , Oxidorreductasas Intramoleculares/metabolismo , Túbulos Renales Proximales/metabolismo , Regeneración , Daño por Reperfusión/metabolismo , Inhibidor Secretorio de Peptidasas Leucocitarias/metabolismo , Factor de Transcripción Activador 4/deficiencia , Factor de Transcripción Activador 4/genética , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Lesión Renal Aguda/prevención & control , Animales , Antígenos de Diferenciación de Linfocitos B/genética , Antígenos de Diferenciación de Linfocitos B/metabolismo , Apoptosis , Autofagia , Hipoxia de la Célula , Línea Celular , Ciclina D1/metabolismo , Modelos Animales de Enfermedad , Factor 2 Eucariótico de Iniciación/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/genética , Túbulos Renales Proximales/patología , Túbulos Renales Proximales/fisiopatología , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Daño por Reperfusión/prevención & control , Inhibidor Secretorio de Peptidasas Leucocitarias/genética , Transducción de Señal , Factores de Tiempo , Transfección
17.
Int J Gynecol Cancer ; 27(4): 634-642, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28338494

RESUMEN

OBJECTIVE: D-dopachrome tautomerase (D-DT) is a homologue of macrophage migration inhibitory factor (MIF) with similar functions. However, the possible biological roles of D-DT in cervical cancer remain unknown so far. METHODS: D-dopachrome tautomerase was assessed by immunohistochemistry in 83 cervical cancer and 31 normal cervix tissues. The stable knockdown of D-DT and MIF by lentivirus-delivered short hairpin RNA was established, and tumor growth was examined in vitro and in vivo. The effects of D-DT and MIF on the migration and invasion were further detected by wound healing assay and transwell assay. Western blot was used to explore the mechanism of D-DT and MIF in cervical cancer pathogenesis. RESULTS: We found that D-DT was significantly high in cervical cancer, which correlated with lymph node metastasis. The knockdown of D-DT and MIF, individually and additively, inhibited the proliferation, migration, and invasion in HeLa and SiHa cells and restrained the growth of xenograft tumor. The ablation of D-DT and MIF rescued the expression of E-cadherin and inhibited the expression of PCNA, cyclin D1, gankyrin, Sam68, and vimentin, as well as phospho-Akt and phospho-glycogen synthase kinase 3-ß. CONCLUSIONS: The inhibition of D-DT and MIF in combination may represent a potential therapeutic strategy for cervical cancer.


Asunto(s)
Oxidorreductasas Intramoleculares/deficiencia , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Neoplasias del Cuello Uterino/metabolismo , Animales , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Regulación hacia Abajo , Femenino , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células HeLa , Xenoinjertos , Humanos , Oxidorreductasas Intramoleculares/biosíntesis , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Metástasis Linfática , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Ratones , Ratones Desnudos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología
18.
Proc Natl Acad Sci U S A ; 113(36): 10198-203, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27551074

RESUMEN

Mutations in superoxide dismutase (SOD1) cause amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disease characterized by the loss of upper and lower motor neurons in the brain and spinal cord. It has been suggested that the toxicity of mutant SOD1 results from its misfolding and accumulation on the cytoplasmic faces of intracellular organelles, including the mitochondria and endoplasmic reticulum (ER) of ALS-affected tissues. Recently, macrophage migration inhibitory factor (MIF) was shown to directly inhibit the accumulation of misfolded SOD1 and its binding to intracellular membranes, but the role of endogenous MIF in modulating SOD1 misfolding in vivo remains unknown. To elucidate this role, we bred MIF-deficient mice with SOD1(G85R) mice, which express a dismutase-inactive mutant of SOD1 and are considered a model of familial ALS. We found that the accumulation of misfolded SOD1, its association with mitochondrial and ER membranes, and the levels of sedimentable insoluble SOD1 aggregates were significantly higher in the spinal cords of SOD1(G85R)-MIF(-/-) mice than in their SOD1(G85R)-MIF(+/+) littermates. Moreover, increasing MIF expression in neuronal cultures inhibited the accumulation of misfolded SOD1 and rescued from mutant SOD1-induced cell death. In contrast, the complete elimination of endogenous MIF accelerated disease onset and late disease progression and shortened the lifespan of the SOD1(G85R) mutant mice. These findings indicate that MIF plays a significant role in the folding and misfolding of SOD1 in vivo, and they have implications for the potential therapeutic role of up-regulating MIF within the nervous system to modulate the selective accumulation of misfolded SOD1.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Mutación , Agregado de Proteínas , Médula Espinal/metabolismo , Superóxido Dismutasa-1/química , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Muerte Celular/genética , Modelos Animales de Enfermedad , Retículo Endoplásmico/química , Retículo Endoplásmico/patología , Femenino , Expresión Génica , Humanos , Oxidorreductasas Intramoleculares/deficiencia , Longevidad/genética , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Masculino , Ratones , Ratones Noqueados , Mitocondrias/química , Mitocondrias/patología , Neuronas/metabolismo , Neuronas/patología , Cultivo Primario de Células , Pliegue de Proteína , Médula Espinal/patología , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo
19.
Arterioscler Thromb Vasc Biol ; 36(1): 108-21, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26543101

RESUMEN

OBJECTIVE: Chronic administration of selective cyclooxygenase-2 (COX-2) inhibitors leads to an increased risk of adverse cardiovascular events, including myocardial infarction and stroke. Vascular smooth muscle cell (VSMC) calcification, a common complication of chronic kidney disease, is directly related to cardiovascular morbidity and mortality. Here, we tested whether specific COX-2 inhibition affects vascular calcification during chronic renal failure. APPROACH AND RESULTS: The COX-2-specific inhibitors NS398 and SC236 significantly increased high-phosphate (Pi)-induced VSMC calcification. Similarly, COX-2(-/-) VSMCs, COX-2(-/-) aortas rings treated with high Pi and adenine diet-induced COX-2(-/-) chronic renal failure mice displayed enhanced calcium deposition. Metabolomic analysis revealed the differential suppression of PGE2 production by COX-1- and COX-2-specific inhibitors in high-Pi-stimulated VSMCs, indicating the involvement of PGE2 during COX-2 inhibition-aggravated vascular calcification. Indeed, exogenous PGE2 reduced alkaline phosphatase activity, osteogenic transdifferentiation, apoptosis, and calcification of VSMCs. In accordance, downregulation of microsomal prostaglandin E synthase (mPGES)-1 in VSMCs, mPGES-1(-/-) aorta with high-Pi stimulation and mPGES-1(-/-) chronic renal failure mice resulted in enhanced vascular mineralization. Further applications of RNAi and specific antagonists for PGE2 receptors indicated EP4 may mediate PGE2-inhibited vascular calcification. CONCLUSIONS: Our data revealed the pivotal role of COX-2-mPGES-1-PGE2 axis in vascular calcification. The selective inhibition of COX-2 or mPGES-1 may increase the risk of calcification and subsequent adverse cardiovascular events during chronic renal failure.


Asunto(s)
Enfermedades de la Aorta/prevención & control , Dinoprostona/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Fallo Renal Crónico/enzimología , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Calcificación Vascular/prevención & control , Adenina , Animales , Aorta Abdominal/enzimología , Aorta Abdominal/patología , Aorta Torácica/enzimología , Aorta Torácica/patología , Enfermedades de la Aorta/inducido químicamente , Enfermedades de la Aorta/enzimología , Enfermedades de la Aorta/genética , Calcitriol , Células Cultivadas , Ciclooxigenasa 2/deficiencia , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/toxicidad , Modelos Animales de Enfermedad , Humanos , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/genética , Fallo Renal Crónico/inducido químicamente , Fallo Renal Crónico/genética , Masculino , Metabolómica/métodos , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Nefrectomía , Fosfatos , Prostaglandina-E Sintasas , Ratas Sprague-Dawley , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Transducción de Señal , Factores de Tiempo , Calcificación Vascular/inducido químicamente , Calcificación Vascular/enzimología , Calcificación Vascular/genética
20.
Thromb Haemost ; 115(1): 200-12, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26310191

RESUMEN

S-nitrosation of macrophage migration inhibitory factor (MIF) has been shown to be cytoprotective in myocardial ischaemia/reperfusion (I/R) injury. Since the exact mechanism of action is unknown, we here characterise the cardioprotective effects of targeted intracellular accumulation of MIF in myocardial I/R injury. We used different in vivo, ex vivo and in vitro models of myocardial I/R and hypoxia/reoxygenation (H/R) injury to determine MIF levels by immunoblots and ELISA in different phases of reperfusion and reoxygenation, respectively. We discovered a rapid decrease of cardiac MIF that was specific to the early phase of reperfusion. Posttranslational modification of MIF via S-nitrosation--proofed by a modified version of the Biotin Switch Assay--prevented this rapid decrease, leading to a targeted intracellular accumulation of MIF in the early phase of reperfusion. Intracellular MIF accumulation preserved the intracellular ability of MIF to reduce oxidative stress as shown by hydrogen peroxide and aconitase activity measurements. Infarct size measurements by TTC staining showed an overall enhanced cardioprotective effect of this protein by reduction of reperfusion injury. In summary, we have unravelled a novel mechanism of MIF-mediated cardioprotection. Targeted intracellular accumulation of MIF by S-nitrosation may offer a novel therapeutic approach in the treatment of myocardial I/R-injury.


Asunto(s)
Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/metabolismo , Aconitato Hidratasa/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Peróxido de Hidrógeno/metabolismo , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Factores Inhibidores de la Migración de Macrófagos/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/patología , Nitrosación , Estrés Oxidativo , Procesamiento Proteico-Postraduccional , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...