Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 41(1): 331-345, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33147994

RESUMEN

OBJECTIVE: Abdominal aortic aneurysm (AAA) is a vascular degenerative disease causing sudden rupture of aorta and significant mortality in elders. Nevertheless, no prognostic and therapeutic target is available for disease management. Gal-1 (galectin-1) is a ß-galactoside-binding lectin constitutively expressed in vasculature with roles in maintaining vascular homeostasis. This study aims to investigate the potential involvement of Gal-1 in AAA progression. Approach and Results: Gal-1 was significantly elevated in circulation and aortic tissues of Ang II (angiotensin II)-infused apoE-deficient mice developing AAA. Gal-1 deficiency reduced incidence and severity of AAA with lower expression of aortic MMPs (matrix metalloproteases) and proinflammatory cytokines. TNFα (tumor necrosis factor alpha) induced Gal-1 expression in cultured vascular smooth muscle cells and adventitial fibroblasts. Gal-1 deletion enhanced TNFα-induced MMP9 expression in fibroblasts but not vascular smooth muscle cells. Cysteinyl-labeling assay demonstrated that aortic Gal-1 exhibited susceptibility to oxidation in vivo. Recombinant oxidized Gal-1 induced expression of MMP9 and inflammatory cytokines to various extents in macrophages, vascular smooth muscle cells, and fibroblasts through activation of MAP (mitogen-activated protein) kinase signaling. Clinically, serum MMP9 level was significantly higher in both patients with AAA and coronary artery disease than in control subjects, whereas serum Gal-1 level was elevated in patients with AAA but not coronary artery disease when compared with controls. CONCLUSIONS: Gal-1 is highly induced and contributes to AAA by enhancing matrix degradation activity and inflammatory responses in experimental model. The pathological link between Gal-1 and AAA is also observed in human patients. These findings support the potential of Gal-1 as a disease biomarker and therapeutic target of AAA.


Asunto(s)
Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/metabolismo , Aortitis/metabolismo , Galectina 1/metabolismo , Remodelación Vascular , Adventicia/metabolismo , Adventicia/patología , Angiotensina II , Animales , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/patología , Aortitis/inducido químicamente , Aortitis/patología , Estudios de Casos y Controles , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Galectina 1/sangre , Galectina 1/deficiencia , Galectina 1/genética , Humanos , Mediadores de Inflamación/metabolismo , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Transducción de Señal , Regulación hacia Arriba
2.
J Biomed Sci ; 28(1): 5, 2021 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-33397354

RESUMEN

BACKGROUND: The accumulation of lipid-laden macrophages, foam cells, within sub-endothelial intima is a key feature of early atherosclerosis. Siglec-E, a mouse orthologue of human Siglec-9, is a sialic acid binding lectin predominantly expressed on the surface of myeloid cells to transduce inhibitory signal via recruitment of SH2-domain containing protein tyrosine phosphatase SHP-1/2 upon binding to its sialoglycan ligands. Whether Siglec-E expression on macrophages impacts foam cell formation and atherosclerosis remains to be established. METHODS: ApoE-deficient (apoE-/-) and apoE/Siglec-E-double deficient (apoE-/-/Siglec-E-/-) mice were placed on high fat diet for 3 months and their lipid profiles and severities of atherosclerosis were assessed. Modified low-density lipoprotein (LDL) uptake and foam cell formation in wild type (WT) and Siglec-E-/-- peritoneal macrophages were examined in vitro. Potential Siglec-E-interacting proteins were identified by proximity labeling in conjunction with proteomic analysis and confirmed by coimmunoprecipitation experiment. Impacts of Siglec-E expression and cell surface sialic acid status on oxidized LDL uptake and signaling involved were examined by biochemical assays. RESULTS: Here we show that genetic deletion of Siglec-E accelerated atherosclerosis without affecting lipid profile in apoE-/- mice. Siglec-E deficiency promotes foam cell formation by enhancing acetylated and oxidized LDL uptake without affecting cholesterol efflux in macrophages in vitro. By performing proximity labeling and proteomic analysis, we identified scavenger receptor CD36 as a cell surface protein interacting with Siglec-E. Further experiments performed in HEK293T cells transiently overexpressing Siglec-E and CD36 and peritoneal macrophages demonstrated that depletion of cell surface sialic acids by treatment with sialyltransferase inhibitor or sialidase did not affect interaction between Siglec-E and CD36 but retarded Siglec-E-mediated inhibition on oxidized LDL uptake. Subsequent experiments revealed that oxidized LDL induced transient Siglec-E tyrosine phosphorylation and recruitment of SHP-1 phosphatase in macrophages. VAV, a downstream effector implicated in CD36-mediated oxidized LDL uptake, was shown to interact with SHP-1 following oxidized LDL treatment. Moreover, oxidized LDL-induced VAV phosphorylation was substantially lower in WT macrophages comparing to Siglec-E-/- counterparts. CONCLUSIONS: These data support the protective role of Siglec-E in atherosclerosis. Mechanistically, Siglec-E interacts with CD36 to suppress downstream VAV signaling involved in modified LDL uptake.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/genética , Antígenos CD36/metabolismo , Células Espumosas/metabolismo , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/farmacología , Animales , Aterosclerosis/metabolismo , Ratones
3.
J Biomed Sci ; 25(1): 6, 2018 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-29361943

RESUMEN

BACKGROUND: Heme oxygenase (HO) catalyzes NADPH-dependent degradation of heme to liberate iron, carbon monoxide and biliverdin. The interaction between HO and cytochrome P450 reductase (CPR), an electron donor, is essential for HO activity. HO-1 is a stress-inducible isoform whereas HO-2 is constitutively expressed. HO-1 induction is commonly seen in cancers and impacts disease progression, supporting the possibility of targeting HO-1 for cancer therapy. METHODS: We employed a cell-based bioluminescence resonance energy transfer assay to screen compounds with ability to inhibit HO-1/CPR interaction. The effect of the identified compound on HO-1/CPR interaction was confirmed by pull down assay. Moreover, the anti-tumorigenic activity of the identified compound on HO-1-enhanced tumor growth and migration was assessed by trypan blue exclusion method and wound healing assay. RESULTS: Danthron was identified as an effective small molecule able to interfere with the interaction between HO-1 and CPR but not HO-2 and CPR. Additional experiments with structural analogues of danthron revealed that the positions of hydroxyl moieties significantly affected the potency of inhibition on HO-1/CPR interaction. Pull-down assay confirmed that danthron inhibited the interaction of CPR with HO-1 but not HO-2. Danthron suppressed growth and migration of HeLa cells with stable HO-1 overexpression but not mock cells. In contrast, anthrarufin, a structural analog with no ability to interfere HO-1/CPR interaction, exhibited no significant effect on HO-1-overexpressing HeLa cells. CONCLUSIONS: These findings demonstrate that danthron is an isoform-specific inhibitor for HO-1/CPR interaction and may serve as a lead compound for novel anticancer drug.


Asunto(s)
Antraquinonas/farmacología , Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Hemo-Oxigenasa 1/metabolismo , NADPH-Ferrihemoproteína Reductasa/metabolismo , Células HEK293 , Células HeLa , Humanos , Isoenzimas/metabolismo
4.
Biochim Biophys Acta ; 1852(11): 2339-51, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26319415

RESUMEN

Endoplasmic reticulum (ER) stress is implicated in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). TRC8 is an ER-resident E3 ligase with roles in modulating lipid and protein biosynthesis. In this study we showed that TRC8 expression was downregulated in steatotic livers of patients and mice fed with a high fat diet (HFD) or a methionine and choline deficient (MCD) diet. To investigate the impact of TRC8 downregulation on steatosis and the progression to non-alcoholic steatohepatitis (NASH), we placed TRC8 knockout (KO) mice and wild type (WT) controls on a HFD or MCD diet and the severities of steatosis and NASH developed were compared. We found that TRC8 deficiency did not significantly affect diet-induced steatosis. Nevertheless, MCD diet-induced NASH as characterized by hepatocyte death, inflammation and fibrosis were exacerbated in TRC8-KO mice. The hepatic ER stress response, as evidenced by increased eIF2α phosphorylation and expression of ATF4 and CHOP, and the level of activated caspase 3, an apoptosis indicator, were augmented by TRC8 deficiency. The hepatic ER stress and NASH induced in mice could be ameliorated by adenovirus-mediated hepatic TRC8 overexpression. Mechanistically, we found that TRC8 deficiency augmented lipotoxic-stress-induced unfolded protein response in hepatocytes by attenuating the arrest of protein translation and the misfolded protein degradation. These findings disclose a crucial role of TRC8 in the maintenance of ER protein homeostasis and its downregulation in steatotic liver contributes to the progression of NAFLD.

5.
Cancer Sci ; 106(3): 299-306, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25580731

RESUMEN

Heme oxygenase-1 (HO-1) is a heme degradation enzyme with antioxidant and immune-modulatory functions. HO-1 promotes tumorigenesis by enhancing tumor cell proliferation and invasion. Whether HO-1 has an effect on cancer progression through stromal compartments is less clear. Here we show that the growth of tumor engrafted subcutaneously in syngeneic mice was not affected by host HO-1 expression. However, lung metastasis arisen from subcutaneous tumor or circulating tumor cells was significantly reduced in HO-1(+/-) mice comparing to wild type (WT) mice. The reduced lung metastasis was also observed in B6 mice bearing HO-1(+/-) bone marrow as comparing to WT chimeras, indicating that HO-1 expression in hematopoietic cells impacts tumor colonization at the metastatic site. Further experiments demonstrated that the numbers of myeloid cells recruited to pulmonary premetastatic niches and metastatic loci were significantly lower in HO-1(+/-) mice than in WT mice. Likewise, the extents of tumor cell extravasation and colonization at the metastatic loci in the early phase of metastasis were significantly lower in HO-1(+/-) mice. Mechanistic studies revealed that HO-1 impacted chemoattractant-induced myeloid cell migration by modulating p38 kinase signaling. Moreover, myeloid HO-1-induced expressions of vascular endothelial growth factor and interleukin-10 promoted tumor cell transendothelial migration and STAT3 activation in vitro. These data support a pathological role of myeloid HO-1 in metastasis and suggest a possibility of targeting myeloid HO-1 for cancer treatment.


Asunto(s)
Proliferación Celular/genética , Transformación Celular Neoplásica/genética , Hemo-Oxigenasa 1/genética , Melanoma Experimental/genética , Proteínas de la Membrana/genética , Invasividad Neoplásica/genética , Animales , Línea Celular Tumoral , Movimiento Celular , Activación Enzimática , Interleucina-10/biosíntesis , Neoplasias Pulmonares/secundario , Masculino , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Transcripción STAT3/metabolismo , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
J Biomed Sci ; 22: 22, 2015 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-25885228

RESUMEN

Heme oxygenase-1 (HO-1) is a rate-limiting enzyme catalyzing oxidative degradation of cellular heme to liberate free iron, carbon monoxide (CO) and biliverdin in mammalian cells. In addition to its primary role in heme catabolism, HO-1 exhibits anti-oxidative and anti-inflammatory functions via the actions of biliverdin and CO, respectively. HO-1 is highly induced in various disease states, including cancer. Several lines of evidence have supported the implication of HO-1 in carcinogenesis and tumor progression. HO-1 deficiency in normal cells enhances DNA damage and carcinogenesis. Nevertheless, HO-1 overexpression in cancer cells promotes proliferation and survival. Moreover, HO-1 induces angiogenesis through modulating expression of angiogenic factors. Although HO-1 is an endoplasmic reticulum resident protein, HO-1 nuclear localization is evident in tumor cells of cancer tissues. It has been shown that HO-1 is susceptible to proteolytic cleavage and translocates to nucleus to facilitate tumor growth and invasion independent of its enzymatic activity. HO-1 also impacts cancer progression through modulating tumor microenvironment. This review summarizes the current understanding of the protumorigenic role of HO-1 and its potential as a molecular target for cancer therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Hemo-Oxigenasa 1/genética , Neoplasias/tratamiento farmacológico , Animales , Hemo-Oxigenasa 1/metabolismo , Humanos , Ratones , Ratas
7.
Arterioscler Thromb Vasc Biol ; 33(4): 785-94, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23393395

RESUMEN

OBJECTIVE: Increased cardiac stromal cell-derived factor-1α (SDF-1α) expression promotes neovascularization and myocardial repair after ischemic injury through recruiting stem cells and reducing cardiomyocyte death. Previous studies have shown that heme oxygenase-1 and its reaction byproduct, carbon monoxide (CO), induce SDF-1α expression in ischemic heart. However, the mechanism underlying heme oxygenase-1/CO-induced cardiac SDF-1α expression remains elusive. This study aims to investigate the signaling pathway and the transcriptional factor that mediate CO-induced SDF-1α gene expression and cardioprotection. APPROACH AND RESULTS: CO gas and a CO-releasing compound, tricarbonyldichlororuthenium (II) dimer, dose-dependently induced SDF-1α expression in primary neonatal cardiomyocytes and H9C2 cardiomyoblasts. Promoter luciferase-reporter assay, electrophoretic mobility shift assay, and chromatin immunoprecipitation demonstrated that the activator protein 2α (AP-2α) mediated tricarbonyldichlororuthenium (II) dimer-induced SDF-1α gene transcription. Tricarbonyldichlororuthenium (II) dimer induced AP-2α expression via protein kinase B (AKT)-dependent signaling. AKT inhibition or AP-2α knockdown reduced tricarbonyldichlororuthenium (II) dimer-induced SDF-1α expression. Coronary ligation induced transient increases of cardiac AP-2α and SDF-1α expression, which were declined at 1 week postinfarction in mice. Periodic exposure of coronary-ligated mice to CO (250 ppm for 1 hour/day, 6 days) resumed the induction of AP-2α and SDF-1α gene expression in infarcted hearts. Immunohistochemistry and echocardiography performed at 4 weeks after coronary ligation revealed that CO treatment enhanced neovascularization in the myocardium of peri-infarct region and improved cardiac function. CO-mediated SDF-1α expression and cardioprotection was ablated by intramyocardial injection of lentivirus bearing specific short hairpin RNA targeting AP-2α. CONCLUSIONS: Our data demonstrate that AKT-dependent upregulation of AP-2α is essential for CO-induced SDF-1α expression and myocardial repair after ischemic injury.


Asunto(s)
Dióxido de Carbono/farmacología , Cardiotónicos/farmacología , Quimiocina CXCL12/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Compuestos Organometálicos/farmacología , Factor de Transcripción AP-2/metabolismo , Administración por Inhalación , Animales , Animales Recién Nacidos , Sitios de Unión , Dióxido de Carbono/administración & dosificación , Dióxido de Carbono/metabolismo , Cardiotónicos/administración & dosificación , Cardiotónicos/metabolismo , Quimiocina CXCL12/genética , Inmunoprecipitación de Cromatina , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ensayo de Cambio de Movilidad Electroforética , Activación Enzimática , Células HeLa , Hemo-Oxigenasa 1/deficiencia , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Hemodinámica/efectos de los fármacos , Humanos , Inmunohistoquímica , Inyecciones Intravenosas , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/patología , Compuestos Organometálicos/administración & dosificación , Compuestos Organometálicos/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Factor de Transcripción AP-2/genética , Transfección , Ultrasonografía , Regulación hacia Arriba , Función Ventricular Izquierda/efectos de los fármacos
8.
J Biol Chem ; 286(5): 3829-38, 2011 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-21115498

RESUMEN

Heme oxygenase-1 (HO-1) is a stress-inducible enzyme catalyzing the oxidative degradation of heme to free iron, CO, and biliverdin. Previous studies demonstrated that HO-1 overexpression promoted VEGF expression and angiogenesis in the ischemic heart. However, the underlying mechanism remained elusive. Here we show that adenovirus-mediated HO-1 transduction of rat primary cardiomyocytes and H9C2 myocytes resulted in significant induction of VEGF expression, and a similar effect was seen in cells directly exposed to CO gas or a CO-releasing compound, tricarbonyldichlororuthenium (II) dimer. HO-1/CO-induced VEGF expression was significantly suppressed by pharmacological inhibition of p38 kinase, but not of AKT, activation. VEGF promoter-luciferase reporter assays, electrophoretic mobility shift assays, supershift assay, and chromatin immunoprecipitation showed that CO-induced VEGF promoter activation requires the binding of the Sp1 transcriptional factor to a cis-regulatory sequence located at the VEGF promoter. Western blot analysis and immunostaining experiments demonstrated that HO-1/CO induced p38-dependent phosphorylation of Sp1 at Thr-453 and Thr-739 both in vitro and in vivo. Overexpression of Sp1 protein with an alanine mutation at Thr-453 or Thr-739 suppressed CO-induced Sp1 binding to the VEGF promoter and its transcriptional activation. Collectively, these data suggest that p38-dependent phosphorylation of Sp1 at Thr-453/Thr-739 is crucial for HO-1/CO-induced VEGF expression in myocytes.


Asunto(s)
Monóxido de Carbono/metabolismo , Hemo-Oxigenasa 1/metabolismo , Células Musculares/metabolismo , Factor de Transcripción Sp1/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Línea Celular , Humanos , Masculino , Miocitos Cardíacos/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Activación Transcripcional
9.
Nat Med ; 8(3): 240-6, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11875494

RESUMEN

The mechanisms underlying the action of the potent anti-inflammatory interleukin-10 (IL-10) are poorly understood. Here we show that, in murine macrophages, IL-10 induces expression of heme oxygenase-1 (HO-1), a stress-inducible protein with potential anti-inflammatory effect, via a p38 mitogen-activated protein kinase-dependent pathway. Inhibition of HO-1 protein synthesis or activity significantly reversed the inhibitory effect of IL-10 on production of tumor necrosis factor-alpha induced by lipopolysaccharide (LPS). Additional experiments revealed the involvement of carbon monoxide, one of the products of HO-1-mediated heme degradation, in the anti-inflammatory effect of IL-10 in vitro. Induction of HO-1 by IL-10 was also evident in vivo. IL-10-mediated protection against LPS-induced septic shock in mice was significantly attenuated by cotreatment with the HO inhibitor, zinc protoporphyrin. The identification of HO-1 as a downstream effector of IL-10 provides new possibilities for improved therapeutic approaches for treating inflammatory diseases.


Asunto(s)
Antiinflamatorios/metabolismo , Hemo Oxigenasa (Desciclizante)/genética , Hemo Oxigenasa (Desciclizante)/metabolismo , Interleucina-10/fisiología , Macrófagos Peritoneales/fisiología , Proteínas Represoras , Factores de Transcripción , Animales , Células Cultivadas , Activación Enzimática , Inducción Enzimática , Inhibidores Enzimáticos/farmacología , Hemo Oxigenasa (Desciclizante)/antagonistas & inhibidores , Hemo-Oxigenasa 1 , Humanos , Interleucina-10/farmacología , Interleucina-6/farmacología , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas de la Membrana , Ratones , Ratones Endogámicos BALB C , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Oligodesoxirribonucleótidos Antisentido/metabolismo , Proteínas/metabolismo , Protoporfirinas/farmacología , Transducción de Señal , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
10.
J Biol Chem ; 284(34): 22672-9, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19556236

RESUMEN

Heme oxygenase-1 (HO-1), a stress-inducible enzyme anchored in the endoplasmic reticulum (ER) by a single transmembrane segment (TMS) located at the C terminus, interacts with NADPH cytochrome P450 reductase and biliverdin reductase to catalyze heme degradation to biliverdin and its metabolite, bilirubin. Previous studies suggested that HO-1 functions as a monomer. Using chemical cross-linking, co-immunoprecipitation, and fluorescence resonance energy transfer (FRET) experiments, here we showed that HO-1 forms dimers/oligomers in the ER. However, oligomerization was not observed with a truncated HO-1 lacking the C-terminal TMS (amino acids 266-285), which exhibited cytosolic and nuclear localization, indicating that the TMS is essential for the self-assembly of HO-1 in the ER. To identify the interface involved in the TMS-TMS interaction, residue Trp-270, predicted by molecular modeling as a potential interfacial residue of TMS alpha-helices, was mutated, and the effects on protein subcellular localization and activity assessed. The results showed that the W270A mutant was present exclusively in the ER and formed oligomers with similar activity to those of the wild type HO-1. Interestingly, the W270N mutant was localized not only in the ER, but also in the cytosol and nucleus, suggesting it is susceptible to proteolytic cleavage. Moreover, the microsomal HO activity of the W270N mutant was significantly lower than that of the wild type. The W270N mutation appears to interfere with the oligomeric state, as revealed by a lower FRET efficiency. Collectively, these data suggest that oligomerization, driven by TMS-TMS interactions, is crucial for the stabilization and function of HO-1 in the ER.


Asunto(s)
Retículo Endoplásmico/enzimología , Hemo-Oxigenasa 1/química , Hemo-Oxigenasa 1/metabolismo , Multimerización de Proteína , Secuencia de Aminoácidos , Línea Celular , Dicroismo Circular , Transferencia Resonante de Energía de Fluorescencia , Hemo-Oxigenasa 1/genética , Humanos , Inmunoprecipitación , Microscopía Confocal , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Homología de Secuencia de Aminoácido
11.
Am J Pathol ; 175(2): 519-32, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19608869

RESUMEN

Oxidative stresses are believed to play an important role in the induction of both cell adhesion molecules and pro-inflammatory cytokines, a key event in a variety of inflammatory processes. The enzyme heme oxygenase-1 (HO-1) functions as an antioxidant and serves to protect against tissue injury. In this study, we report that HO-1 was induced in cultured human tracheal smooth muscle cells after either treatment with a potent inducer of HO-1 activity, cobalt protoporphyrin IX, or infection with a recombinant adenovirus that carries the human HO-1 gene. Overexpression of HO-1 protected against tumor necrosis factor (TNF)-alpha-mediated airway inflammation via the down-regulation of oxidative stress, adhesion molecules, and interleukin-6 in both cultured human tracheal smooth muscle cells and the airways of mice. In addition, HO-1 overexpression inhibited TNF-alpha-induced intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 expression, adherence of THP-1 cells, generation of interleukin-6, p47(phox) translocation, and nuclear factor-kappaB activation. HO-1 overexpression also attenuated TNF-alpha-induced oxidative stress, which was abrogated in the presence of both the HO-1 inhibitor, zinc protoporphyrin IX, as well as a carbon monoxide scavenger. In addition, HO-1 overexpression reduced the formation of a TNFR1/c-Src/p47(phox) complex. These results suggest that HO-1 functions as a suppressor of TNF-alpha signaling, not only by inhibiting the expression of adhesion molecules and generation of interleukin-6, but also by diminishing intracellular reactive oxygen species production and nuclear factor-kappaB activation in both cultured human tracheal smooth muscle cells and the airways of mice.


Asunto(s)
Hemo-Oxigenasa 1/biosíntesis , Músculo Liso/inmunología , Estrés Oxidativo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Traqueítis/inmunología , Animales , Moléculas de Adhesión Celular/biosíntesis , Línea Celular , Regulación hacia Abajo , Humanos , Ratones , Músculo Liso/enzimología , Músculo Liso/patología , NADPH Oxidasas/metabolismo , FN-kappa B/metabolismo , Protoporfirinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Traqueítis/enzimología , Traqueítis/patología , Factor de Necrosis Tumoral alfa/farmacología , Familia-src Quinasas/metabolismo
12.
J Biomed Sci ; 17: 12, 2010 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-20175935

RESUMEN

BACKGROUND: The microsatellite polymorphism of heme oxygenase (HO)-1 gene promoter has been shown to be associated with the susceptibility to ischemic event, including coronary artery disease (CAD), myocardial infarction, and peripheral vascular disease. We aimed to examine whether the length of (GT)(n) repeats in HO-1 gene promoter is associated with ischemic stroke in people with CAD risk factors, especially low level of HDL. METHODS: A total of 183 consecutive firstever ischemic stroke inpatients and 164 non-stroke patients were screened for the length of (GT)(n) repeats in HO-1 promoter. The long (L) and short (S) genotype are defined as the averaged repeat number >26 and <==26, respectively. RESULTS: Stroke patients tended to have more proportions of hypertension, diabetics and genotype L, than those of genotype S. Patients with genotype L of HO-1 gene promoter have higher stroke risk in comparison with genotype S especially in dyslipidemia individuals. The significant differences on stroke risk in multivariate odds ratios were found especially in people with low HDL-C levels. CONCLUSIONS: Subjects carrying longer (GT)(n) repeats in HO-1 gene promoter may have greater susceptibility to develop cerebral ischemic only in the presence of low HDL-C, suggesting the protective effects in HO-1 genotype S in the process of ischemic stroke, particularly in subjects with poor HDL-C status.


Asunto(s)
Dislipidemias/genética , Hemo-Oxigenasa 1/genética , Polimorfismo Genético , Regiones Promotoras Genéticas , Accidente Cerebrovascular/genética , HDL-Colesterol/sangre , HDL-Colesterol/metabolismo , Dislipidemias/complicaciones , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Genotipo , Guanosina/química , Humanos , Masculino , Factores de Riesgo , Accidente Cerebrovascular/etiología , Tiamina/química
13.
Biochim Biophys Acta ; 1783(10): 1826-34, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18544348

RESUMEN

The present study investigated the cellular mechanism underlying the degradation of heme oxygenase-1 (HO-1), an endoplasmic reticulum (ER)-anchored protein. The turnover of HO-1 induced in vascular smooth muscle cells (VSMCs) was significantly attenuated by proteasome inhibitors, suggesting the involvement of a proteasome-mediated pathway. High molecular weight ubiquitin conjugates were co-immunoprecipitated with HO-1 from VSMCs after proteasome inhibition, and HO-1 ubiquitination was confirmed in HEK293 cells overexpressing His-tagged HO-1 and HA-tagged ubiquitin. Endogenous p97, an ATPase, and Ufd1, both implicated as essential components in the ER-associated degradation pathway (ERAD), were co-eluted with His-tagged HO-1 from metal affinity resin. Knockdown of either p97 or Ufd1 in HEK293 cells using specific siRNA significantly prolonged the half-life of endogenously induced HO-1 and slowed the degradation of ubiquitinated HO-1. HO-1 ubiquitination in HEK293 cells was enhanced by zinc chloride, but suppressed with a zinc chelator (N,N,N',N'-tetrakis(2-pyridylmethyl)ethyl-enediamine), suggesting the involvement of a RING-E3 ligase in this process. Collectively, these data indicate that HO-1 protein turnover is regulated by the ubiquitin-proteasome system through the ERAD pathway.


Asunto(s)
Retículo Endoplásmico/enzimología , Hemo-Oxigenasa 1/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Animales , Células Cultivadas , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/metabolismo , Hemo-Oxigenasa 1/genética , Humanos , Lisosomas/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/enzimología , Inhibidores de Proteasas/farmacología , Transporte de Proteínas , ARN Interferente Pequeño/genética , Ratas
14.
J Cell Physiol ; 219(3): 617-25, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19170060

RESUMEN

Increased neovascularization is commonly observed in hemorrhagic plaques and associated with rupture of atherosclerotic lesions. This study aims to investigate whether hemin accumulated at the site of hematoma promotes neovascularization through affecting the growth and function of endothelial progenitor cells (EPCs) and the possible mechanism involved. Here we demonstrated that hemin promoted a significant increase in neovessel formation in matrigel plugs embedded in vivo and enhanced the proliferation and endothelial gene expression in EPCs in vitro. VEGF-induced migration response and the capability to incorporate into the vascular networks were markedly enhanced in hemin-treated EPCs. Hemin induced the phosphorylation of ERK and AKT but not p38 or JNK. The inhibition of AKT or ERK activation significantly attenuated the effect of hemin on cell proliferation. However, the enhanced migration response induced by hemin was significantly suppressed by the inhibition of AKT but not ERK. Hemin induced significant increase in reactive oxygen species (ROS) production and hemin-induced angiogenic response of EPCs was substantially reduced by treatment with N-acetylcysteine. Collectively, these data support that hemin-induced ROS mediates the activation of AKT and ERK signaling pathways, which in turn promotes the cell proliferation and function of EPCs. J. Cell. Physiol. 219: 617-625, 2009. (c) 2009 Wiley-Liss, Inc.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Hemina/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/enzimología , Activación Enzimática/efectos de los fármacos , Células Madre Hematopoyéticas/enzimología , Hemo-Oxigenasa 1/deficiencia , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
15.
Oncogene ; 38(10): 1688-1701, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30348988

RESUMEN

Signal peptide peptidase (SPP) is an endoplasmic reticulum (ER)-resident aspartyl protease mediating intramembrane cleavage of type II transmembrane proteins. Increasing evidence has supported the role of SPP in ER-associated protein degradation. In the present study, we show that SPP expression is highly induced in human lung and breast cancers and correlated with disease outcome. Stable depletion of SPP expression in lung and breast cancer cell lines significantly reduced cell growth and migration/invasion abilities. Quantitative analysis of the proteomic changes of microsomal proteins in lung cancer cells by the stable isotope labeling with amino acids in cell culture (SILAC) approach revealed that the level of FKBP8, an endogenous inhibitor of mTOR, was significantly increased following SPP depletion. Co-immunoprecipitation assay and confocal immunofluorescence demonstrated that SPP interacted and colocalized with FKBP8 in ER, supporting that FKBP8 is a protein substrate of SPP. Cycloheximide chase and proteasome inhibition experiments revealed that SPP-mediated proteolysis facilitated FKBP8 protein degradation in cytosol. Further experiment demonstrated that the levels of phosphorylation in mTOR and its downstream effectors, S6K and 4E-BP1, were significantly lower in SPP-depleted cells. The reduced mTOR signaling and decreases of growth and migration/invasion abilities induced by SPP depletion in cancer cells could be reversed by FKBP8 downregulation. The implication of FKBP8 in SPP-mediated tumorigenicity was also observed in the xenograft model. Together, these findings disclose that SPP promotes tumor progression, at least in part, via facilitating the degradation of FKBP8 to enhance mTOR signaling.


Asunto(s)
Ácido Aspártico Endopeptidasas/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Regulación hacia Arriba , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Neoplasias Pulmonares/patología , Células MCF-7 , Ratones , Trasplante de Neoplasias , Proteolisis , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
16.
J Mol Cell Cardiol ; 45(1): 44-55, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18534615

RESUMEN

Heme oxygenase-1 (HO-1) is a stress-inducible enzyme with multiple protective functions in cardiovascular systems. Studies have shown that the timely cardiac HO-1 overexpression at acute phase of ischemic infarction (MI) provides protection via its anti-apoptotic and anti-inflammatory effects. Here we demonstrate that a delayed HO-1 transduction mediated by a recombinant adeno-associated virus in ischemic hearts of mice with permanent coronary artery ligation significantly attenuated left ventricular fibrosis and cardiac dysfunctions examined at 4 weeks post MI. HO-1-mediated protection was correlated with enhanced vascularization in the ischemic myocardium. HO-1 gene transfer resulted in a notable increase in the number of c-kit(+)- stem cells recruited to the infarcted area at 10 days after ligation. HO-1-mediated stem cell recruitment was also demonstrated in the heart of non-ischemic mice receiving intravenous infusion of green fluorescent protein-bearing bone marrow stem cells. Additional experiments revealed that vascular endothelial growth factor (VEGF) and stromal cell-derived factor-1 (SDF-1) were highly induced in HO-1 transduced myocardium. Mononuclear cell infiltration was evident and colocalized with angiogenic factors in the same region. Flow cytometry analysis of the mononuclear cells isolated from HO-1-transduced left ventricles revealed that over 50% of cells expressed CD34, a marker of hematopoietic stem cells and endothelial progenitor cells. VEGF and SDF-1 blockade by neutralizing antibodies significantly attenuated HO-1-mediated neovascularization and protection in infarcted mice. These data suggest that cardiac HO-1 gene transfer post MI provides protection at least in part by promoting neovascularization through inducing angiogenic factors and the recruitment of circulating progenitor/stem cells.


Asunto(s)
Quimiocina CXCL12/biosíntesis , Hemo-Oxigenasa 1/biosíntesis , Infarto del Miocardio/enzimología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Animales , Antígenos CD34/metabolismo , Quimiocina CXCL12/genética , Dependovirus , Terapia Genética , Ventrículos Cardíacos/enzimología , Células Madre Hematopoyéticas/enzimología , Hemo-Oxigenasa 1/genética , Leucocitos Mononucleares/enzimología , Masculino , Ratones , Infarto del Miocardio/genética , Infarto del Miocardio/terapia , Miocardio/enzimología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factores de Tiempo , Transducción Genética , Factor A de Crecimiento Endotelial Vascular/genética
17.
Sci Rep ; 8(1): 11497, 2018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-30065303

RESUMEN

Vascular smooth muscle cell (VSMC) migration play a key role in the development of intimal hyperplasia and atherosclerosis. Galectin-1 (Gal-1) is a redox-sensitive ß-galactoside-binding lectin expressed in VSMCs with intracellular and extracellular localizations. Here we show that VSMCs deficient in Gal-1 (Gal-1-KO) exhibited greater motility than wild type (WT) cells. Likewise, Gal-1-KO-VSMC migration was inhibited by a redox-insensitive but activity-preserved Gal-1 (CSGal-1) in a glycan-dependent manner. Gal-1-KO-VSMCs adhered slower than WT cells on fibronectin. Cell spreading and focal adhesion (FA) formation examined by phalloidin and vinculin staining were less in Gal-1-KO-VSMCs. Concomitantly, FA kinase (FAK) phosphorylation was induced to a lower extent in Gal-1-KO cells. Analysis of FA dynamics by nocodazole washout assay demonstrated that FA disassembly, correlated with FAK de-phosphorylation, was faster in Gal-1-KO-VSMCs. Surface plasmon resonance assay demonstrated that CSGal-1 interacted with α5ß1integrin and fibronectin in a glycan-dependent manner. Chemical crosslinking experiment and atomic force microscopy further revealed the involvement of extracellular Gal-1 in strengthening VSMC-fibronectin interaction. In vivo experiment showed that carotid ligation-induced neointimal hyperplasia was more severe in Gal-1-KO mice than WT counterparts. Collectively, these data disclose that Gal-1 restricts VSMC migration by modulating cell-matrix interaction and focal adhesion turnover, which limits neointimal formation post vascular injury.


Asunto(s)
Benzamidas/metabolismo , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Adhesiones Focales/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Tirosina/análogos & derivados , Animales , Células Cultivadas , Fibronectinas/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Adhesiones Focales/patología , Humanos , Hiperplasia/metabolismo , Hiperplasia/patología , Integrina alfa5beta1/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Neointima/metabolismo , Neointima/patología , Fosforilación/fisiología , Ratas , Ratas Sprague-Dawley , Tirosina/metabolismo
18.
Adv Healthc Mater ; 5(20): 2686-2697, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27592617

RESUMEN

In patients who survive myocardial infarction, many go on to develop congestive heart failure (CHF). Despite ongoing efforts to develop new approaches for postinfarction therapy, there are still no effective therapeutic options available to CHF patients. Currently, the delivery of cardioprotective drugs relies entirely on passive uptake via the enhanced permeability and retention (EPR) effect which occurs in proximity to the infarction site. However, in ischemic disease, unlike in cancer, the EPR effect only exists for a short duration postinfarction and thus insufficient for meaningful cardioprotection. Splenic monocytes are recruited to the heart in large numbers postinfarction, and are known to interact with platelets during circulation. Therefore, the strategy is to exploit this interaction by developing platelet-like proteoliposomes (PLPs), biomimicking platelet interactions with circulating monocytes. PLPs show strong binding affinity for monocytes but not for endothelial cells in vitro, mimicking normal platelet activity. Furthermore, intravital multiphoton imaging shows that comparing to plain liposomes, PLPs do not aggregate on uninjured endothelium but do accumulate at the injury site 72 h postinfarction. Importantly, PLPs enhance the targeting of anti-inflammatory drug, cobalt protoporphyrin, to the heart in an EPR-independent manner, which result in better therapeutic outcome.


Asunto(s)
Materiales Biomiméticos/administración & dosificación , Plaquetas/química , Células Endoteliales/efectos de los fármacos , Corazón/efectos de los fármacos , Monocitos/efectos de los fármacos , Infarto del Miocardio/terapia , Cicatrización de Heridas/efectos de los fármacos , Animales , Materiales Biomiméticos/química , Cardiotónicos/administración & dosificación , Cardiotónicos/química , Línea Celular , Humanos , Inflamación/tratamiento farmacológico , Liposomas/química , Masculino , Ratones , Ratones Endogámicos BALB C , Permeabilidad , Activación Plaquetaria/efectos de los fármacos , Células RAW 264.7
19.
Circulation ; 110(3): 309-16, 2004 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-15226216

RESUMEN

BACKGROUND: Heme oxygenase-1 (HO-1) is a stress-response enzyme implicated in cardioprotection. To explore whether HO-1 has a role in cardiac remodeling response, the effect of its overexpression on angiotensin II (Ang II)-induced cardiac hypertrophy was examined. METHODS AND RESULTS: HO-1 was induced in cultured rat neonatal cardiomyocytes by treatment with cobalt protoporphyrin IX (CoPPIX) or a recombinant adenovirus carrying the human HO-1 gene. Ang II-induced myocyte hypertrophy assessed by increments in cell size, [3H]leucine uptake, and protein content was suppressed by HO-1 overexpression. Cotreatment of cells with tin protoporphyrin IX, a HO inhibitor, significantly reversed the suppressive effect of HO-1. Bilirubin, one of the byproducts of heme degradation by HO-1, mediated the suppressive effect through the inhibition of Ang II-induced production of reactive oxygen species, as detected by a 2',7'-dichlorofluorescein probe. The antihypertrophic effect of HO-1 was also demonstrated in rats receiving chronic Ang II infusions. Cotreatment of animals with CoPPIX significantly attenuated Ang II-induced left ventricular hypertrophy and hyperdynamic contractions, whereas concomitant treatment with tin protoporphyrin IX abolished CoPPIX-mediated cardioprotection in vivo. CONCLUSIONS: HO-1 attenuates Ang II-induced cardiac hypertrophy both in vitro and in vivo, and bilirubin mediates, at least in part, the antihypertrophic effect of HO-1 via inhibition of reactive oxygen species production after Ang II stimulation.


Asunto(s)
Angiotensina II/antagonistas & inhibidores , Cardiomegalia/inducido químicamente , Hemo Oxigenasa (Desciclizante)/fisiología , Animales , Bilirrubina/fisiología , Cardiomegalia/metabolismo , Cardiomegalia/patología , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1 , Masculino , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo
20.
J Ocul Pharmacol Ther ; 21(6): 420-8, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16386083

RESUMEN

AIMS: The aim of this study was to investigate the efficiency of adenoviral-mediated transfer of the interleukin (IL)-10 gene for inhibition of experimental autoimmune anterior uveitis, a rat model of human acute anterior uveitis. Uveitis was induced in the Lewis rat by simultaneous injection of melanin-associated antigen intraperitoneally (i.p.) and into the left footpad. The animals were treated by systemic administration of adenoviral construct expressing IL-10 (Ad-IL-10) or Ad-Mock carrying no cytokine transgene. RESULTS: A significant reduction in ocular inflammation was noted for rats that received one or two divided i.p. administrations of Ad-IL-10 (one 10 x 10(9) and two 5 x 10(9) particles of adenoviral construct, respectively), as judged by reduced clinical scores and decreased leukocyte infiltration in the anterior chamber and confirmed by histological examinations, relative to control animals. Systemic Ad-IL-10, treatment also revealed a higher serum level of IL-10, compared to the controls. CONCLUSIONS: The results suggest that systemic adenovirus-mediated IL-10 gene therapy has an anti-inflammatory effect on immune-mediated ocular inflammation and that this approach may be promising for the treatment of acute anterior uveitis.


Asunto(s)
Adenoviridae , Enfermedades Autoinmunes/terapia , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Interleucina-10/genética , Uveítis Anterior/terapia , Adenoviridae/genética , Animales , Cámara Anterior/inmunología , Cámara Anterior/patología , Humor Acuoso/inmunología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Quimiotaxis de Leucocito/inmunología , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Interleucina-10/biosíntesis , Interleucina-10/sangre , Recuento de Leucocitos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Reacción en Cadena de la Polimerasa , Ratas , Ratas Endogámicas Lew , Factor de Necrosis Tumoral alfa/biosíntesis , Uveítis Anterior/inmunología , Uveítis Anterior/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA