Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 136(6): 749-754, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32548640

RESUMEN

Several studies demonstrate that hemolysis and free heme in circulation cause endothelial barrier dysfunction and are associated with severe pathological conditions such as acute respiratory distress syndrome, acute chest syndrome, and sepsis. However, the precise molecular mechanisms involved in the pathology of heme-induced barrier disruption remain to be elucidated. In this study, we investigated the role of free heme in the endothelial barrier integrity and mechanisms of heme-mediated intracellular signaling of human lung microvascular endothelial cells (HLMVECs). Heme, in a dose-dependent manner, induced a rapid drop in the endothelial barrier integrity of HLMVECs. An investigation into barrier proteins revealed that heme primarily affected the tight junction proteins zona occludens-1, claudin-1, and claudin-5, which were significantly reduced after heme exposure. The p38MAPK/HSP27 pathway, involved in the regulation of endothelial cytoskeleton remodeling, was also significantly altered after heme treatment, both in HLMVECs and mice. By using a knockout (KO) mouse for MKK3, a key regulator of the p38MAPK pathway, we showed that this KO effectively decreased heme-induced endothelial barrier dysfunction. Taken together, our results indicate that targeting the p38MAPK pathway may represent a crucial treatment strategy in alleviating hemolytic diseases.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Hemo/farmacología , MAP Quinasa Quinasa 3/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Animales , Antígenos CD/análisis , Cadherinas/análisis , Permeabilidad Capilar/fisiología , Células Cultivadas , Claudinas/análisis , Células Endoteliales/fisiología , Proteínas de Choque Térmico HSP27/fisiología , Proteínas de Choque Térmico/fisiología , Hemólisis , Humanos , Pulmón/irrigación sanguínea , MAP Quinasa Quinasa 3/deficiencia , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Noqueados , Microvasos/citología , Chaperonas Moleculares/fisiología , Uniones Estrechas/efectos de los fármacos , Proteína de la Zonula Occludens-1/análisis , Proteínas Quinasas p38 Activadas por Mitógenos
2.
Arch Biochem Biophys ; 641: 50-61, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29407793

RESUMEN

Myoclonic epilepsy with ragged-red fibers (MERRF) is a maternally inherited mitochondrial neuromuscular disease. We previously reported a significant decrease of mRNA and protein levels of nuclear DNA-encoded carbonic anhydrase VIII (CA8) in MERRF cybrids harboring A8344G mutation in mitochondrial DNA (mtDNA). In this study, we established a reporter construct of luciferase gene-carrying hCA8 promoter containing several putative transcription factor-binding sites, including GC-box, AP-2 and TATA-binding element in the 5'flanking region of the hCA8 gene. Using a series of mutated hCA8 promoter constructs, we demonstrated that a proximal GC-box, recognized by Sp1 and other Sp family members, may be a key cis-element functioning at the promoter. Additionally, a significant increase of the hCA8 promoter activity was observed in the wild-type and mutant cybrids with over-expression of eGFP-Sp1, but no detectable increase in the CA8 protein expression. In contrast, over-expression of Flag-Sp1 and Flag-Sp4 significantly increased the hCA8 promoter activity as well as endogenous CA8 protein expression in neuron-like HEK-293 T cells. However, down-regulation of Sp1, but not Sp4, in 293 T cells revealed a significant reduction of CA8 expression, suggesting that Sp1 is a predominant transcription factor for regulation of CA8 activity. Furthermore, our data indicate that chromatin structure may be involved in the expression of hCA8 gene in MERRF cybrids. Taken together, these results suggest that Sp1 transactivates hCA8 gene through the proximal GC box element in the promoter region. The key modulator-responsive factor to the mtDNA mutation and how it may affect nuclear hCA8 gene transcription need further investigations.


Asunto(s)
Biomarcadores de Tumor/genética , Regulación Enzimológica de la Expresión Génica , Síndrome MERRF/enzimología , Modelos Biológicos , Regiones Promotoras Genéticas , Transcripción Genética , Sitios de Unión , ADN Mitocondrial/genética , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Proteínas de Choque Térmico HSP27/fisiología , Humanos , Síndrome MERRF/genética , Mutación , Factores de Transcripción/metabolismo
3.
J Physiol ; 595(14): 4857-4873, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28481416

RESUMEN

KEY POINTS: Muscular contractions performed using a combination of low external loads and partial restriction of limb blood flow appear to induce substantial gains in muscle strength and muscle mass. This exercise regime may initially induce muscular stress and damage; however, the effects of a period of blood flow restricted training on these parameters remain largely unknown. The present study shows that short-term, high-frequency, low-load muscle training performed with partial blood flow restriction does not induce significant muscular damage. However, signs of myocellular stress and inflammation that were observed in the early phase of training and after the training intervention, respectively, may be facilitating the previously reported gains in myogenic satellite cell content and muscle hypertrophy. The present results improve our current knowledge about the physiological effects of low-load muscular contractions performed under blood flow restriction and may provide important information of relevance for future therapeutic treatment of muscular atrophy. ABSTRACT: Previous studies indicate that low-load muscle contractions performed under local blood flow restriction (BFR) may initially induce muscle damage and stress. However, whether these factors are evoked with longitudinal BFR training remains unexplored at the myocellular level. Two distinct study protocols were conducted, covering 3 weeks (3 wk) or one week (1 wk). Subjects performed BFR exercise (100 mmHg, 20% 1RM) to concentric failure (BFRE) (3 wk/1 wk), while controls performed work-matched (LLE) (3 wk) or high-load (HLE; 70% 1RM) (1 wk) free-flow exercise. Muscle biopsies (3 wk) were obtained at baseline (Pre), 8 days into the intervention (Mid8), and 3 and 10 days after training cessation (Post3, Post10) to examine macrophage (M1/M2) content as well as heat shock protein (HSP27/70) and tenascin-C expression. Blood samples (1 wk) were collected before and after (0.1-24 h) the first and last training session to examine markers of muscle damage (creatine kinase), oxidative stress (total antibody capacity, glutathione) and inflammation (monocyte chemotactic protein-1, interleukin-6, tumour necrosis factor α). M1-macrophage content increased 108-165% with BFRE and LLE at Post3 (P < 0.05), while M2-macrophages increased (163%) with BFRE only (P < 0.01). Membrane and intracellular HSP27 expression increased 60-132% at Mid8 with BFRE (P < 0.05-0.01). No or only minor changes were observed in circulating markers of muscle damage, oxidative stress and inflammation. The amplitude, timing and localization of the above changes indicate that only limited muscle damage was evoked with BFRE. This study is the first to show that a period of high-frequency, low-load BFR training does not appear to induce general myocellular damage. However, signs of tissue inflammation and focal myocellular membrane stress and/or reorganization were observed that may be involved in the adaptation processes evoked by BFR muscle exercise.


Asunto(s)
Ejercicio Físico/fisiología , Proteínas de Choque Térmico HSP27/fisiología , Proteínas HSP70 de Choque Térmico/fisiología , Macrófagos/fisiología , Músculo Esquelético/fisiología , Flujo Sanguíneo Regional , Adulto , Quimiocina CCL2/sangre , Creatina Quinasa/sangre , Humanos , Interleucina-6/sangre , Masculino , Músculo Esquelético/irrigación sanguínea , Mialgia , Percepción del Dolor , Factor de Necrosis Tumoral alfa/sangre , Regulación hacia Arriba , Adulto Joven
4.
Klin Onkol ; 29(1): 29-38, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26879061

RESUMEN

Heat shock proteins (HSPs) HSP27, HSP70 and HSP90 are molecular chaperones; their expression is increased after exposure of cells to conditions of environmental stress, including heat shock, heavy metals, oxidative stress, or pathologic conditions, such as ischemia, infection, and inflammation. Their protective function is to help the cell cope with lethal conditions. The HSPs are a class of proteins which, in normal cells, are responsible for maintaining homeostasis, interacting with diverse protein substrates to assist in their folding, and preventing the appearance of folding intermediates that lead to misfolded or damaged molecules. They have been shown to interact with different key apoptotic proteins and play a crucial role in regulating apoptosis. Several HSPs have been demonstrated to directly interact with various components of tightly regulated caspase-dependent programmed cell death. These proteins also affect caspase-independent apoptosis by interacting with apoptogenic factors. Heat shock proteins are aberrantly expressed in hematological malignancies. Because of their prognostic implications and functional role in leukemias, HSPs represent an interesting target for antileukemic therapy. This review will describe different molecules interacting with anti-apoptotic proteins HSP70 and HSP90, which can be used in cancer therapy based on their inhibition.


Asunto(s)
Proteínas de Choque Térmico/fisiología , Leucemia/patología , Apoptosis , Proteínas de Choque Térmico HSP27/fisiología , Proteínas HSP70 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP70 de Choque Térmico/química , Proteínas HSP70 de Choque Térmico/fisiología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/química , Proteínas HSP90 de Choque Térmico/fisiología , Humanos
5.
Int J Cancer ; 136(6): E496-507, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25130271

RESUMEN

Increased expression of the molecular chaperone Hsp27 is associated with the progression of prostate cancer (PCa) to castration-resistant disease, which is lethal due to metastatic spread of the prostate tumor. Metastasis requires epithelial to mesenchymal transition (EMT), which endows cancer cells with the ability to disseminate from the primary tumor and colonize new tissue sites. A wide variety of secreted factors promote EMT, and while overexpression and constitutive activation of epidermal growth factor (EGF) signaling is associated with poor prognosis of PCa, a precise role of EGF in PCa progression to metastasis remains unclear. Here, we show that Hsp27 is required for EGF-induced cell migration, invasion and MMPs activity as well as the expression of EMT markers including Fibronectin, Vimentin and Slug with concomitant decrease of E-cadherin. Mechanistically, we found that Hsp27 is required for EGF-induced AKT and GSK3ß phosphorylation and ß-catenin nuclear translocation. Moreover, silencing Hsp27 decreases EGF dependent phosphorylation of ß-catenin on tyrosine 142 and 654, enhances ß-catenin ubiquitination and degradation, prevents ß-catenin nuclear translocation and binding to the Slug promoter. These data suggest that Hsp27 is required for EGF-mediated EMT via modulation of the ß-catenin/Slug signaling pathway. Together, our findings underscore the importance of Hsp27 in EGF induced EMT in PCa and highlight the use of Hsp27 knockdown as a useful strategy for patients with advanced disease.


Asunto(s)
Factor de Crecimiento Epidérmico/fisiología , Transición Epitelial-Mesenquimal , Proteínas de Choque Térmico HSP27/fisiología , Neoplasias de la Próstata/patología , beta Catenina/fisiología , Transporte Activo de Núcleo Celular , Línea Celular Tumoral , Movimiento Celular , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Proteínas de Choque Térmico , Humanos , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Chaperonas Moleculares , Invasividad Neoplásica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
6.
World J Urol ; 33(3): 323-7, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24798454

RESUMEN

PURPOSE: Androgen receptor (AR) networks are predominantly involved in prostate cancer (PCa) progression; consequently, factors of AR regulation represent promising targets for PCa therapy. The ErbB3-binding protein 1 (Ebp1) is linked to AR suppression and chemoresistance by so far unknown mechanisms. In this study, an assumed regulation of Ebp1 by the newly identified AR controlling signaling axis heat-shock protein 27 (HSP27)-microRNA-1 (miR-1) was examined. METHODS: Transfection experiments were carried out overexpressing and knockdown HSP27 and miR-1, respectively, in LNCaP and PC-3 cells. Afterward, HSP27- and miR-1-triggered Ebp1 protein expression was monitored by Western blotting. RESULTS: AR-positive LNCaP cells and AR-negative PC-3 cells possessed diverse basal expression levels of Ebp1. However, subsequent studies revealed no differences in cellular Ebp1 concentrations after modulation of HSP27 and miR-1. Furthermore, docetaxel incubation experiments exhibited no effects on Ebp1 protein synthesis. CONCLUSION: In PCa, Ebp1 has been described as a regulator of AR functionality and as an effector of PCa therapy resistance. Our data suggest that Ebp1 functionality is independent from heat-shock-protein-regulated progression networks in PCa.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Adenocarcinoma/fisiopatología , Proteínas de Choque Térmico HSP27/fisiología , MicroARNs/fisiología , Neoplasias de la Próstata/fisiopatología , Proteínas de Unión al ARN/fisiología , Receptores Androgénicos/fisiología , Transducción de Señal/fisiología , Adenocarcinoma/patología , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/fisiología , Células Cultivadas , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Masculino , Neoplasias de la Próstata/patología , Receptores Androgénicos/efectos de los fármacos , Transfección
7.
Am J Physiol Heart Circ Physiol ; 306(12): H1714-9, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24727494

RESUMEN

Omentin is a novel adipocytokine expressed in visceral adipose tissue. Secretion and blood concentration of omentin decrease in the obese subjects. We previously demonstrated that omentin is anti-inflammatory in vascular smooth muscle cells (SMCs). While vascular remodeling via migration of SMCs is also important for hypertension development, it remains to be clarified whether omentin affects this process. Here we examined whether omentin controls SMC migration. Omentin (300 ng/ml, 2 h) significantly inhibited platelet-derived growth factor (PDGF)-BB (10 ng/ml, 6 h)-induced migration of rat mesenteric arterial SMCs, as determined by Boyden chamber assay. Omentin (300 ng/ml, 2 h) significantly inhibited PDGF-BB (10 ng/ml, 30 min)-induced phosphorylation of p38 and heat shock protein (HSP) 27. Omentin (300 ng/ml, 2 h) significantly inhibited PDGF-BB (10 ng/ml, 30 min)-induced NADPH oxidase (NOX) activation as determined by lucigenin assay. Omentin (300 ng/ml, 24 h) significantly inhibited fetal bovine serum (5%, 4 days)-induced SMC outgrowth from rat isolated mesenteric artery. In vivo, omentin significantly inhibited carotid intimal hyperplasia in mouse ligation model. In summary, we for the first time demonstrate that omentin prevents PDGF-BB-induced SMC migration by preventing NOX/O2(-)/p38/HSP27 pathways, which might be at least partly responsible for the preventive effects on neointimal hyperplasia. Our data suggest that omentin may be protective against hypertension development by inhibiting vascular structural remodeling.


Asunto(s)
Adipoquinas/farmacología , Antioxidantes/fisiología , Movimiento Celular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Proteínas Proto-Oncogénicas c-sis/farmacología , Transducción de Señal/fisiología , Adipoquinas/fisiología , Animales , Becaplermina , Arterias Carótidas/citología , Arterias Carótidas/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Citocinas/farmacología , Proteínas Ligadas a GPI , Proteínas de Choque Térmico HSP27/fisiología , Técnicas In Vitro , Lectinas/farmacología , Masculino , Arterias Mesentéricas/citología , Arterias Mesentéricas/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Modelos Animales , Miocitos del Músculo Liso/citología , NADPH Oxidasas/fisiología , Oxígeno/fisiología , Proteínas Proto-Oncogénicas c-sis/fisiología , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
8.
Chin J Physiol ; 57(6): 329-34, 2014 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-25575521

RESUMEN

In view of the cardioprotective effect of oxytocin (OT) released in response to stress, the aim of this study was to evaluate the role of heat shock proteins Hsps 70, 27 and 20 in stress-induced cardioprotection in isolated, perfused rat hearts. Rats were divided in two main groups: unstressed and stressed rats, and all of them were subjected to i.c.v. infusion of vehicle or drugs: unstressed rats [control: vehicle, OT (100 ng/5 µl), atosiban (ATO; 4.3 µg/5 µl) as OT antagonist, ATO+OT], and stressed rats [St: stress, OT+St, ATO+St]. After anesthesia, hearts were isolated and subjected to 30 min regional ischemia and 60 min subsequent reperfusion (IR). Acute stress protocol included swimming for 10 min before anesthesia. Malondialdehyde in coronary effluent was measured and the expression of Hsp 70, 27 and 20 was measured in myocardium using real-time reverse transcriptase polymerase chain reaction (RT-PCR). The malondialdehyde levels, which decreased in the St and OT groups, increased by the administration of atosiban as an OT antagonist. The expression of Hsp27 increased 4 to 5 folds by stress induction and i.c.v. infusion of OT. Central administration of atosiban prior to both stress and OT decreased Hsp27 mRNA levels. These findings suggest that endogenous OT may participate in stress-induced cardioprotection via Hsp27 over-expression as an early response.


Asunto(s)
Proteínas de Choque Térmico HSP27/fisiología , Daño por Reperfusión Miocárdica/prevención & control , Oxitocina/fisiología , Estrés Fisiológico , Animales , Masculino , Malondialdehído/análisis , Oxitocina/antagonistas & inhibidores , Ratas , Ratas Wistar , Regulación hacia Arriba
9.
J Oral Pathol Med ; 42(1): 9-16, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22563860

RESUMEN

Photodynamic therapy (PDT) of cells is a new treatment modality involving selective delivery of a photosensitive dye into target cells, followed by visible light irradiation. PDT induces cell death by excessive ROS generation. The effects of multiple photosensitizers were owing to the difference in cell types involving sensitizer-specific protein changes linked to resistance. HSP27 is regulated in response to stress and is associated with apoptotic process. The effects of HSP27 on PDT resistance are controversial and unclear. The purpose of this study was to investigate the role of HSP27 down-regulation in the PDT-induced cells and HSP27 regulation in the resistance to PDT. KB cells transfected with HSP27 siRNA were exposed to hematoporphyrin (HP) followed by irradiation at 635 nm at an energy density of 4.5 mW/cm(2). After irradiation, the effects on HSP27 down-regulation were assessed by MTT assay, flow cytometry, confocal analysis, Western blotting and caspase activity. The results of this study showed that down-regulation of HSP27 restored cell survival in HP-PDT-induced apoptotic KB cells. HSP27 down-regulation attenuated PDT-induced apoptosis through caspase-mediated pathway in KB cells. Also, HSP27 silencing regulated Bax, Bcl-2, and PARP protein expression in PDT-induced cells. Therefore, HSP27 down-regulation confers resistance to PDT through interruption of apoptotic protein activity in PDT-induced cell death. HSP27 might contribute to regulating PDT-induced apoptosis in PDT-resistant cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Choque Térmico HSP27 , Neoplasias de la Boca/tratamiento farmacológico , Fotoquimioterapia , Caspasas/genética , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Silenciador del Gen , Proteínas de Choque Térmico HSP27/genética , Proteínas de Choque Térmico HSP27/fisiología , Humanos , Células KB , Neoplasias de la Boca/enzimología , Neoplasias de la Boca/genética , ARN Interferente Pequeño
10.
J Neurosci ; 31(33): 11992-2000, 2011 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-21849559

RESUMEN

More than 80% of Alzheimer's disease (AD) patients have some degree of cerebral amyloid angiopathy (CAA). In addition to arteries and veins, capillaries can also be affected. Capillary CAA (capCAA), rather than CAA in larger vessels, is associated with flame-like amyloid-beta (Aß) deposits that may extend beyond the vessel wall and radiate into the neuropil, a phenomenon also known as "dyshoric angiopathy." Aß deposits in AD, parenchymal as well as (cap)CAA and dyshoric angiopathy, are associated with a local inflammatory reaction, including activation of microglial cells and astrocytes that, among others, produce cytokines and reactive oxygen species. This neuroinflammatory reaction may account for at least part of the cognitive decline. In previous studies we observed that small heat shock proteins (sHsps) are associated with Aß deposits in AD. In this study the molecular chaperones Hsp20, HspB8 and HspB2B3 were found to colocalize with CAA and capCAA in AD brains. In addition, Hsp20, HspB8 and HspB2B3 colocalized with intercellular adhesion molecule 1 (ICAM-1) in capCAA-associated dyshoric angiopathy. Furthermore, we demonstrated that Hsp20, HspB8 and HspB2B3 induced production of interleukin 8, soluble ICAM-1 and monocyte chemoattractant protein 1 by human leptomeningeal smooth muscle cells and human brain astrocytes in vitro and that Hsp27 inhibited production of transforming growth factor beta 1 and CD40 ligand. Our results suggest a central role for sHsps in the neuroinflammatory reaction in AD and CAA and thus in contributing to cognitive decline.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Encéfalo/metabolismo , Encéfalo/patología , Angiopatía Amiloide Cerebral/metabolismo , Angiopatía Amiloide Cerebral/patología , Proteínas de Choque Térmico Pequeñas/fisiología , Mediadores de Inflamación/fisiología , Anciano , Anciano de 80 o más Años , Astrocitos/metabolismo , Astrocitos/patología , Células Cultivadas , Femenino , Proteínas del Choque Térmico HSP20/fisiología , Proteínas de Choque Térmico HSP27/fisiología , Proteínas de Choque Térmico/fisiología , Humanos , Masculino , Chaperonas Moleculares , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Recombinantes/farmacología
11.
Biochemistry ; 51(28): 5695-702, 2012 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-22734719

RESUMEN

Heat shock proteins (HSPs) play a critical role in many intracellular processes, including apoptosis and delivery of other proteins to intracellular compartments. Small HSPs have been shown previously to participate in many cellular functions, including IL-8 induction. Human adenovirus infection activates intracellular signaling, involving particularly the c-Src and mitogen-activated protein kinases [Natarajan, K., et al. (2003) J. Immunol. 170, 6234-6243]. HSP27 and MK2 are also phosphorylated, and c-Src, and its downstream targets, p38, ERK1/2, and c-Jun-terminal kinase (JNK), differentially mediate IL-8 and MCP-1 expression. Specifically, activation and translocation of transcription factor NFκB-p65 occurs in a p38-dependent fashion [Rajaiya, J., et al. (2009) Mol. Vision 15, 2879-2889]. Herein, we report a novel role for HSP27 in an association of p38 with NFκB-p65. Immunoprecipitation assays of virus-infected but not mock-infected cells revealed a signaling complex including p38 and NFκB-p65. Transfection with HSP27 short interfering RNA (siRNA) but not scrambled RNA disrupted this association and reduced the level of IL-8 expression. Transfection with HSP27 siRNA also reduced the level of nuclear localization of NFκB-p65 and p38. By use of tagged p38 mutants, we found that amino acids 279-347 of p38 are necessary for the association of p38 with NFκB-p65. These studies strongly suggest that HSP27, p38, and NFκB-p65 form a signalosome in virus-infected cells and influence downstream expression of pro-inflammatory mediators.


Asunto(s)
Infecciones por Adenovirus Humanos/metabolismo , Adenovirus Humanos/fisiología , Proteínas de Choque Térmico HSP27/fisiología , Infecciones por Adenovirus Humanos/virología , Células Cultivadas , Quimiocinas/biosíntesis , Humanos , Queratinocitos/virología , Subunidad p50 de NF-kappa B/metabolismo , Transporte de Proteínas , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Lab Invest ; 92(1): 32-45, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21931298

RESUMEN

Apoptosis is a driving force of diabetic end-organ damage, including diabetic nephropathy (DN). However, the mechanisms that modulate diabetes-induced cell death are not fully understood. Heat shock protein 27 (HSP27/HSPB1) is a cell stress protein that regulates apoptosis in extrarenal cells and is expressed by podocytes exposed to toxins causing nephrotic syndrome. We investigated the regulation of HSPB1 expression and its function in podocytes exposed to factors contributing to DN, such as high glucose and angiotensin (Ang) II. HSPB1 expression was assessed in renal biopsies from patients with DN, minimal change disease or focal segmental glomerulosclerosis (FSGS), in a rat model of diabetes induced by streptozotocin (STZ) and in Ang II-infused rats. The regulation of HSPB1 was studied in cultured human podocytes and the function of HSPB1 expressed in response to pathophysiologically relevant stimuli was explored by short interfering RNA knockdown. Total kidney HSPB1 mRNA and protein expression was increased in rats with STZ-induced diabetes and in rats infused with Ang II. Upregulation of HSPB1 protein was confirmed in isolated diabetic glomeruli. Immunohistochemistry showed increased glomerular expression of HSPB1 in both models and localized glomerular HSPB1 to podocytes. HSPB1 protein was increased in glomerular podocytes from patients with DN or FSGS. In cultured human podocytes HSPB1 mRNA and protein expression was upregulated by high glucose concentrations and Ang II. High glucose, but not Ang II, promoted podocyte apoptosis. HSPB1 short interfering RNA (siRNA) targeting increased apoptosis in a high-glucose milieu and sensitized to Ang II or TGFß1-induced apoptosis by promoting caspase activation. In conclusion, both high glucose and Ang II contribute to HSPB1 upregulation. HSPB1 upregulation allows podocytes to better withstand an adverse high-glucose or Ang II-rich environment, such as can be found in DN.


Asunto(s)
Angiotensina II/farmacología , Apoptosis , Glucosa/farmacología , Proteínas de Choque Térmico HSP27/fisiología , Podocitos/fisiología , Adaptación Fisiológica , Adolescente , Adulto , Anciano , Animales , Caspasas/metabolismo , Células Cultivadas , Niño , Preescolar , Nefropatías Diabéticas/metabolismo , Proteínas de Choque Térmico HSP27/análisis , Proteínas de Choque Térmico , Humanos , Persona de Mediana Edad , Chaperonas Moleculares , Ratas , Ratas Endogámicas WKY
13.
J Biomed Sci ; 19: 52, 2012 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-22621211

RESUMEN

BACKGROUND: Autophagy and molecular chaperones both regulate protein homeostasis and maintain important physiological functions. Atg7 (autophagy-related gene 7) and Hsp27 (heat shock protein 27) are involved in the regulation of neurodegeneration and aging. However, the genetic connection between Atg7 and Hsp27 is not known. METHODS: The appearances of the fly eyes from the different genetic interactions with or without polyglutamine toxicity were examined by light microscopy and scanning electronic microscopy. Immunofluorescence was used to check the effect of Atg7 and Hsp27 knockdown on the formation of autophagosomes. The lifespan of altered expression of Hsp27 or Atg7 and that of the combination of the two different gene expression were measured. RESULTS: We used the Drosophila eye as a model system to examine the epistatic relationship between Hsp27 and Atg7. We found that both genes are involved in normal eye development, and that overexpression of Atg7 could eliminate the need for Hsp27 but Hsp27 could not rescue Atg7 deficient phenotypes. Using a polyglutamine toxicity assay (41Q) to model neurodegeneration, we showed that both Atg7 and Hsp27 can suppress weak, toxic effect by 41Q, and that overexpression of Atg7 improves the worsened mosaic eyes by the knockdown of Hsp27 under 41Q. We also showed that overexpression of Atg7 extends lifespan and the knockdown of Atg7 or Hsp27 by RNAi reduces lifespan. RNAi-knockdown of Atg7 expression can block the extended lifespan phenotype by Hsp27 overexpression, and overexpression of Atg7 can extend lifespan even under Hsp27 knockdown by RNAi. CONCLUSIONS: We propose that Atg7 acts downstream of Hsp27 in the regulation of eye morphology, polyglutamine toxicity, and lifespan in Drosophila.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila , Ojo , Proteínas de Choque Térmico HSP27/genética , Animales , Autofagia/genética , Proteína 7 Relacionada con la Autofagia , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Epistasis Genética/genética , Ojo/anatomía & histología , Ojo/crecimiento & desarrollo , Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Choque Térmico HSP27/fisiología , Péptidos/toxicidad , Interferencia de ARN
14.
J Immunol ; 184(10): 5582-8, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20385876

RESUMEN

Differences in CD8(+)CD57(-) and CD8(+)CD57(+) lymphocyte lifespan have been documented. Lower numbers and shorter lifespan are characteristic of CD8(+)CD57(+) in normal individuals. However, CD8(+)CD57(+) are expanded in certain disease states including T cell large granular leukemia and other hematologic malignancies. The mechanisms responsible for the differences in CD8(+)CD57(-) and CD8(+)CD57(+) lifespan remain elusive. In this study, we demonstrate that the small heat shock protein (Hsp) 27 is a key regulator of CD8(+)CD57(+) lymphocyte lifespan. We found that Hsp27 expression is significantly lower in CD8(+)CD57(+) than in CD8(+)CD57(-) lymphocytes. In contrast, Hsp60 and Hsp70 are expressed at comparable levels. Unlike other antiapoptotic Bcl-2-like molecules, the expression of Hsp27 tightly correlates with CD8(+)CD57(+) and CD8(+)CD57(-) lifespan. We demonstrate that Hsp27 overexpression in CD8(+)CD57(+) lymphocytes to levels found normally in CD8(+)CD57(-) lymphocytes decreased apoptosis. Accordingly, silencing of Hsp27 in CD8(+)CD57(-) lymphocytes increased apoptosis. Collectively these results demonstrate that Hsp27 is a critical regulator of normal CD8(+)CD57(+) lifespan supporting its use as a marker of lifespan in this lineage, and suggest a mechanism responsible for the decreased apoptosis and clonal expansion characteristic of certain disease states.


Asunto(s)
Antígenos CD57/biosíntesis , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proteínas de Choque Térmico HSP27/fisiología , Apoptosis/genética , Apoptosis/inmunología , Antígenos CD57/genética , Antígenos CD57/metabolismo , Linfocitos T CD8-positivos/citología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Células Cultivadas , Células Clonales , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Regulación de la Expresión Génica/inmunología , Marcadores Genéticos/genética , Proteínas de Choque Térmico HSP27/biosíntesis , Proteínas de Choque Térmico HSP27/genética , Humanos , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
15.
Hum Cell ; 35(1): 238-249, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34791597

RESUMEN

Ferroptosis, as an new form of non-apoptotic regulated cell death, plays an important role in human cancers. Although it is reported that HSP27 is an novel regulator of ferroptosis in cancer, it remains unknown how HSP27 affects ferroptosis in glioma. In this study, we examined the effect of HSP27 on the ferroptosis of glioblasotma. HSP27 overexpression protects glioblastoma cells from erastin-induced ferroptosis while HSP27 depletion promotes erastin-induced ferroptosis of glioblastoma. Notably, HSP27 phosphorylation is required for the protective function of HSP27 in erastin-induced ferroptosis. Overall, our study reveal novel molecular mechanisms of ferroptosis in glioma and also identify HSP27 as a negative regulator of ferroptosis and a potential target for the treatment of glioma.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Ferroptosis/genética , Glioblastoma/genética , Glioblastoma/patología , Proteínas de Choque Térmico HSP27/fisiología , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Ferroptosis/fisiología , Expresión Génica/genética , Glioblastoma/terapia , Proteínas de Choque Térmico HSP27/genética , Proteínas de Choque Térmico HSP27/metabolismo , Humanos , Terapia Molecular Dirigida , Fosforilación , Piperazinas
16.
J Neurosci ; 30(46): 15374-82, 2010 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-21084594

RESUMEN

Molecular chaperones regulate the aggregation of a number of proteins that pathologically misfold and accumulate in neurodegenerative diseases. Identifying ways to manipulate these proteins in disease models is an area of intense investigation; however, the translation of these results to the mammalian brain has progressed more slowly. In this study, we investigated the ability of one of these chaperones, heat shock protein 27 (Hsp27), to modulate tau dynamics. Recombinant wild-type Hsp27 and a genetically altered version of Hsp27 that is perpetually pseudo-phosphorylated (3×S/D) were generated. Both Hsp27 variants interacted with tau, and atomic force microscopy and dynamic light scattering showed that both variants also prevented tau filament formation. However, extrinsic genetic delivery of these two Hsp27 variants to tau transgenic mice using adeno-associated viral particles showed that wild-type Hsp27 reduced neuronal tau levels, whereas 3×S/D Hsp27 was associated with increased tau levels. Moreover, rapid decay in hippocampal long-term potentiation (LTP) intrinsic to this tau transgenic model was rescued by wild-type Hsp27 overexpression but not by 3×S/D Hsp27. Because the 3×S/D Hsp27 mutant cannot cycle between phosphorylated and dephosphorylated states, we can conclude that Hsp27 must be functionally dynamic to facilitate tau clearance from the brain and rescue LTP; however, when this property is compromised, Hsp27 may actually facilitate accumulation of soluble tau intermediates.


Asunto(s)
Proteínas de Choque Térmico HSP27/fisiología , Simulación de Dinámica Molecular , Plasticidad Neuronal/genética , Proteínas tau/genética , Proteínas tau/metabolismo , Animales , Femenino , Hipocampo/metabolismo , Hipocampo/patología , Masculino , Ratones , Ratones Transgénicos , Fosforilación/fisiología
17.
J Virol ; 84(7): 3654-65, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20106928

RESUMEN

Stress granules are sites of mRNA storage formed in response to a variety of stresses, including viral infections. Here, the mechanisms and consequences of stress granule formation during poliovirus infection were examined. The results indicate that stress granules containing T-cell-restricted intracellular antigen 1 (TIA-1) and mRNA are stably constituted in infected cells despite lacking intact RasGAP SH3-domain binding protein 1 (G3BP) and eukaryotic initiation factor 4G. Fluorescent in situ hybridization revealed that stress granules in infected cells do not contain significant amounts of viral positive-strand RNA. Infection does not prevent stress granule formation in response to heat shock, indicating that poliovirus does not block de novo stress granule formation. A mutant TIA-1 protein that prevents stress granule formation during oxidative stress also prevents formation in infected cells. However, stress granule formation during infection is more dependent upon ongoing transcription than is formation during oxidative stress or heat shock. Furthermore, Sam68 is recruited to stress granules in infected cells but not to stress granules formed in response to oxidative stress or heat shock. These results demonstrate that stress granule formation in poliovirus-infected cells utilizes a transcription-dependent pathway that results in the appearance of stable, compositionally unique stress granules.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , Poliovirus/fisiología , Factor 2 Eucariótico de Iniciación/metabolismo , Proteínas de Choque Térmico HSP27/fisiología , Células HeLa , Proteínas de Choque Térmico , Respuesta al Choque Térmico , Humanos , Chaperonas Moleculares , Fosforilación , Proteínas de Unión a Poli(A)/análisis , Proteínas de Unión a Poli(A)/fisiología , ARN Mensajero/biosíntesis , Antígeno Intracelular 1 de las Células T , Transcripción Genética
18.
Hepatology ; 51(5): 1614-23, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20209605

RESUMEN

UNLABELLED: miR-17-5p is overexpressed in hepatocellular carcinoma (HCC), but the specific regulatory mechanisms of miR-17-5p in HCC remain unknown. We investigated the molecular basis of miR-17-5p as an oncogene in human HCC cell lines. Our in vivo and in vitro data indicate that miR-17-5p up-regulates the migration and proliferation of HCC cells. Interestingly, proteomic and western blotting assays revealed that miR-17-5p significantly activates the p38 mitogen-activated protein kinase MAPK pathway and increases the phosphorylation of heat shock protein 27 (HSP27). Our results also suggest that E2F1-dependent down-regulation of Wip1 regulates miR-17-5p-p38-HSP27 signaling. Furthermore, suppression of HSP27 expression by small interfering RNA or the p38 MAPK pathway-specific inhibitor SB203580 decreases the migration of HCC cells overexpressing miR-17-5p but does not reduce their proliferation. Finally, we show that miR-17-5p expression correlates well with HSP27 status in primary human HCC tissues with metastasis. CONCLUSION: Our findings suggest that the p38 MAPK pathway plays a crucial role in miR-17-5p-induced phosphorylation of HSP27 and, as a consequence, phosphorylated HSP27 enhances the migration of HCC cells. Our data highlight an important role of miR-17-5p in the proliferation and migration of HCC cells and support the potential application of miR-17-5p in HCC therapy.


Asunto(s)
Proteínas de Choque Térmico HSP27/fisiología , MicroARNs/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Factor de Transcripción E2F1/fisiología , Proteínas de Choque Térmico HSP27/biosíntesis , Humanos , Imidazoles/farmacología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Fosfoproteínas Fosfatasas/fisiología , Proteína Fosfatasa 2C , Piridinas/farmacología , ARN Interferente Pequeño/farmacología
19.
J Immunol ; 182(1): 675-83, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19109201

RESUMEN

Proteomic analysis of murine skin has shown that a variety of heat shock proteins (HSPs) are constitutively expressed in the skin. Using murine allergic contact hypersensitivity as a model, we investigated the role of two heat shock proteins, HSP27 and HSP70, in the induction of cutaneous cell-mediated immune responses. Immunohistochemical examination of skin specimens showed that HSP27 was present in the epidermis and HSP70 was present in both the epidermis and dermis. Inhibition of HSP27 and HSP70 produced a reduction in the 1-fluoro-2,4-dinitrobenzene contact hypersensitivity response and resulted in the induction of Ag-specific unresponsiveness. Treatment of dendritic cell cultures with recombinant HSP27 caused in the up-regulation of IL-1beta, TNF-alpha, IL-6, IL-12p70, and IL-12p40 but not IL-23p19, which was inhibited when Abs to HSP27 were added. The 1-fluoro-2,4-dinitrobenzene-conjugated dendritic cells that had been treated with HSP27 had an increased capacity to initiate contact hypersensitivity responses compared with control dendritic cells. This augmented capacity required TLR4 signaling because neither cytokine production by dendritic cells nor the increased induction of contact hypersensitivity responses occurred in TLR4-deficient C3H/HeJ mice. Our findings indicate that a cascade of events occurs following initial interaction of hapten with the skin that includes increased activity of HSPs, their interaction with TLR4, and, in turn, increased production of cytokines that are known to enhance Ag presentation by T cells. The results suggest that HSPs form a link between adaptive and innate immunity during the early stages of contact hypersensitivity.


Asunto(s)
Dermatitis Alérgica por Contacto/inmunología , Dermatitis Alérgica por Contacto/metabolismo , Proteínas de Choque Térmico HSP27/fisiología , Proteínas HSP70 de Choque Térmico/fisiología , Piel/química , Piel/inmunología , Administración Tópica , Animales , Anticuerpos/administración & dosificación , Presentación de Antígeno , Células Cultivadas , Citocinas/biosíntesis , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Dermatitis Alérgica por Contacto/prevención & control , Dinitrofluorobenceno/administración & dosificación , Dinitrofluorobenceno/inmunología , Femenino , Proteínas de Choque Térmico HSP27/antagonistas & inhibidores , Proteínas de Choque Térmico HSP27/inmunología , Proteínas HSP70 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP70 de Choque Térmico/inmunología , Haptenos/administración & dosificación , Haptenos/inmunología , Inmunidad Celular , Inmunohistoquímica , Ratones , Ratones Endogámicos C3H , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética
20.
Anticancer Res ; 41(3): 1171-1181, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33788708

RESUMEN

BACKGROUND/AIM: We have previously reported the identification of the cytotoxic chemotype compound-I (CC-I) from a chemical library screening against glioblastoma. MATERIALS AND METHODS: The biological activity of CC-I on drug-resistant neuroblastomas [e.g., HFE gene variant C282Y stably transfected human neuroblastoma SH-SY5Y cells (C282Y HFE/SH-SY5Y), SK-N-AS] was characterized using cell culture models and in vivo mouse tumor models. RESULTS: CC-I had potent cytotoxicity on therapy-resistant neuroblastoma cells and limited cytotoxicity on human primary dermal fibroblast cells. In addition, CC-I showed a robust anti-tumor effect on therapy-resistant human neuroblastoma C282Y HFE/SH-SY5Y cells but not on SK-N-AS cells in a subcutaneous tumor model. CC-I induced phosphorylation of heat shock protein 27 (HSP27), protein kinase B (Akt), and c-Jun N-terminal kinase (JNK) in C282Y HFE/SH-SY5Y neuroblastoma cells. CONCLUSION: CC-I may be an effective therapeutic option for therapy-resistant neuroblastomas, especially if they express the C282Y HFE gene variant. Its anti-tumor effects are possibly through HSP27-Akt-JNK activation.


Asunto(s)
Antineoplásicos/farmacología , Neuroblastoma/tratamiento farmacológico , Tiobarbitúricos/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Niño , Preescolar , Femenino , Fibroblastos/efectos de los fármacos , Proteínas de Choque Térmico HSP27/fisiología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Masculino , Ratones , Neuroblastoma/patología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/fisiología , Tiobarbitúricos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA