Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Int J Mol Sci ; 22(8)2021 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-33923911

RESUMEN

Human small heat shock proteins are molecular chaperones that regulate fundamental cellular processes in normal and pathological cells. Here, we have reviewed the role played by HspB1, HspB4 and HspB5 in the context of Cystic Fibrosis (CF), a severe monogenic autosomal recessive disease linked to mutations in Cystic Fibrosis Transmembrane conductance Regulator protein (CFTR) some of which trigger its misfolding and rapid degradation, particularly the most frequent one, F508del-CFTR. While HspB1 and HspB4 favor the degradation of CFTR mutants, HspB5 and particularly one of its phosphorylated forms positively enhance the transport at the plasma membrane, stability and function of the CFTR mutant. Moreover, HspB5 molecules stimulate the cellular efficiency of currently used CF therapeutic molecules. Different strategies are suggested to modulate the level of expression or the activity of these small heat shock proteins in view of potential in vivo therapeutic approaches. We then conclude with other small heat shock proteins that should be tested or further studied to improve our knowledge of CFTR processing.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/genética , Animales , Cristalinas/genética , Proteínas de Choque Térmico/genética , Humanos , Chaperonas Moleculares/genética , Mutación , Cadena B de alfa-Cristalina/genética
2.
J Cell Sci ; 125(Pt 5): 1141-51, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22302993

RESUMEN

We previously found that the NF-κB transcription factor is activated during the recovery period after heat shock; moreover, we demonstrated that NF-κB is essential for cell survival after heat shock by activating autophagy, a mechanism that probably helps the cell to cope with hyperthermic stress through clearance of damaged proteins. In this study, we analyze the involvement of NF-κB in basal and heat-stress-induced protein quality control, by comparing the level of multiubiquitylated and/or aggregated proteins, and proteasome and autophagic activity in NF-κB-competent and NF-κB-incompetent cells. We show that NF-κB has only a minor role in basal protein quality control, where it modulates autophagosome maturation. By contrast, NF-κB is shown to be a key player in protein quality control after hyperthermia. Indeed, NF-κB-incompetent cells show highly increased levels of multiubiquitylated and/or aggregated proteins and aggresome clearance defects; a phenotype that disappears when NF-κB activity is restored to normal. We demonstrate that during heat shock recovery NF-κB activates selective removal of misfolded or aggregated proteins--a process also called 'aggrephagy'--by controlling the expression of BAG3 and HSPB8 and by modulating the level of the BAG3-HspB8 complex. Thus NF-κB-mediated increase in the level of the BAG3-HspB8 complex leads to upregulation of aggrephagy and clearance of irreversibly damaged proteins and might increase cell survival in conditions of hyperthermia.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autofagia/fisiología , Proteínas de Choque Térmico/metabolismo , Respuesta al Choque Térmico/fisiología , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Transcripción ReIA/metabolismo , Proteínas Reguladoras de la Apoptosis , Línea Celular Tumoral , Supervivencia Celular , Células HeLa , Humanos , Chaperonas Moleculares , FN-kappa B/genética , Pliegue de Proteína , Factor de Transcripción ReIA/deficiencia , Factor de Transcripción ReIA/genética , Ubiquitinación
3.
Int J Hyperthermia ; 29(5): 409-22, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23697380

RESUMEN

PURPOSE: The recent discoveries in the field of human small heat shock proteins (sHSPs) clearly point to the important roles played by these adenosine triphosphate (ATP)-independent chaperones in the regulation of a large spectrum of vital cellular processes and in pathological diseases. These proteins are therefore considered as very attractive therapeutic targets. AIMS: To understand the functions of the stress-inducible members of the sHSP family, HspB1, HspB5 and HspB8, and be able to therapeutically modulate their activities, researchers are faced with the complex oligomerisation and phosphorylation properties of these proteins and with their ability to interact with each other and with specific protein targets. Here, we have integrated, in a functionally orientated way, the up-to-date literature data concerning HspB1, HspB5 and HspB8 protein interactions which reflect their numerous crucial cellular functions. We also present data supporting the idea that specific phospho-oligomeric domains of HspB1 are involved in the interaction with particular client proteins. CONCLUSIONS: More information concerning the interactions between client protein targets and sHSPs or the multiple combinatorial chimeric oligomeric complexes formed by different sHSPs are urgently required to elaborate a comprehensive sHSPs protein interactome and propose efficient and pathology-specific therapeutic approaches.


Asunto(s)
Proteínas de Choque Térmico HSP27/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Cadena B de alfa-Cristalina/metabolismo , Animales , Humanos , Chaperonas Moleculares , Transducción de Señal
4.
Arch Toxicol ; 87(1): 19-48, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22885793

RESUMEN

Heat shock proteins (HSP) are a subset of the molecular chaperones, best known for their rapid and abundant induction by stress. HSP genes are activated at the transcriptional level by heat shock transcription factor 1 (HSF1). During the progression of many types of cancer, this heat shock transcriptional regulon becomes co-opted by mechanisms that are currently unclear, although evidently triggered in the emerging tumor cell. Concerted activation of HSF1 and the accumulation of HSPs then participate in many of the traits that permit the malignant phenotype. Thus, cancers of many histologies exhibit activated HSF1 and increased HSP levels that may help to deter tumor suppression and evade therapy in the clinic. We review here the extensive work that has been carried out and is still in progress aimed at (1) understanding the oncogenic mechanisms by which HSP genes are switched on, (2) determining the roles of HSF1/HSP in malignant transformation and (3) discovering approaches to therapy based on disrupting the influence of the HSF1-controlled transcriptome in cancer.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Choque Térmico/fisiología , Neoplasias/genética , Neoplasias/patología , Factores de Transcripción/genética , Chaperonina 60/genética , Chaperonina 60/metabolismo , Proteínas de Unión al ADN/metabolismo , Resistencia a Antineoplásicos , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Factores de Transcripción del Choque Térmico , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/radioterapia , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Transducción de Señal , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
5.
Exp Cell Res ; 316(9): 1535-52, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20233592

RESUMEN

Human HspB1 (also denoted Hsp27) is an oligomeric anti-apoptotic protein that has tumorigenic and metastatic roles. To approach the structural organizations of HspB1 that are active in response to apoptosis inducers acting through different pathways, we have analyzed the relative protective efficiency induced by this protein as well its localization, oligomerization and phosphorylation. HeLa cells, that constitutively express high levels of HspB1 were treated with either etoposide, Fas agonist antibody, staurosporine or cytochalasin D. Variability in HspB1 efficiency to interfere with the different apoptotic transduction pathways induced by these agents were detected. Moreover, inducer-specific dynamic changes in HspB1 localization, native size and phosphorylation were observed, that differed from those observed after heat shock. Etoposide and Fas treatments gradually shifted HspB1 towards large but differently phosphorylated oligomeric structures. In contrast, staurosporine and cytochalasin D induced the rapid but transient formation of small oligomers before large structures were formed. These events correlated with inducer-specific phosphorylations of HspB1. Of interest, the formation of small oligomers in response to staurosporine and cytochalasin D was time correlated with the rapid disruption of F-actin. The subsequent, or gradual in the case of etoposide and Fas, formation of large oligomeric structures was a later event concomitant with the early phase of caspase activation. These observations support the hypothesis that HspB1 has the ability, through specific changes in its structural organization, to adapt and interfere at several levels with challenges triggered by different signal transduction pathways upstream of the execution phase of apoptosis.


Asunto(s)
Apoptosis/fisiología , Proteínas de Choque Térmico HSP27/metabolismo , Transducción de Señal , Actinas/metabolismo , Antineoplásicos Fitogénicos/farmacología , Western Blotting , Caspasa 3/metabolismo , Citocalasina D/farmacología , Citocromos c/metabolismo , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Etopósido/farmacología , Técnica del Anticuerpo Fluorescente , Células HeLa , Proteínas de Choque Térmico , Respuesta al Choque Térmico , Humanos , Mitocondrias/efectos de los fármacos , Chaperonas Moleculares , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Fosforilación/efectos de los fármacos , Estaurosporina/farmacología , Receptor fas/metabolismo
6.
Cancer Res ; 80(10): 1970-1980, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32060146

RESUMEN

The Sonic Hedgehog (SHH) pathway plays a key role in cancer. Alterations of SHH canonical signaling, causally linked to tumor progression, have become rational targets for cancer therapy. However, Smoothened (SMO) inhibitors have failed to show clinical benefit in patients with cancers displaying SHH autocrine/paracrine expression. We reported earlier that the SHH receptor Patched (PTCH) is a dependence receptor that triggers apoptosis in the absence of SHH through a pathway that differs from the canonical one, thus generating a state of dependence on SHH for survival. Here, we propose a dual function for SHH: its binding to PTCH not only activates the SHH canonical pathway but also blocks PTCH-induced apoptosis. Eighty percent, 64%, and 8% of human colon, pancreatic, and lung cancer cells, respectively, overexpressed SHH at transcriptional and protein levels. In addition, SHH-overexpressing cells expressed all the effectors of the PTCH-induced apoptotic pathway. Although the canonical pathway remained unchanged, autocrine SHH interference in colon, pancreatic, and lung cell lines triggered cell death through PTCH proapoptotic signaling. In vivo, SHH interference in colon cancer cell lines decreased primary tumor growth and metastasis. Therefore, the antitumor effect associated to SHH deprivation, usually thought to be a consequence of the inactivation of the canonical SHH pathway, is, at least in part, because of the engagement of PTCH proapoptotic activity. Together, these data strongly suggest that therapeutic strategies based on the disruption of SHH/PTCH interaction in SHH-overexpressing cancers should be explored. SIGNIFICANCE: Sonic Hedgehog-overexpressing tumors express PTCH-induced cell death effectors, suggesting that this death signaling could be activated as an antitumor strategy.


Asunto(s)
Apoptosis/fisiología , Proteínas Hedgehog/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Receptores Patched/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/fisiología , Embrión de Pollo , Xenoinjertos , Humanos , Ratones , Transducción de Señal/fisiología , Pez Cebra
7.
Int J Radiat Oncol Biol Phys ; 70(2): 543-53, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17980509

RESUMEN

PURPOSE: The ability of heat shock protein 27 (Hsp27) to protect cells from stressful stimuli and its increased levels in tumors resistant to anticancer therapeutics suggest that it may represent a target for sensitization to radiotherapy. In this study, we investigate the protective role of Hsp27 against radiation-induced apoptosis and the effect of its attenuation in highly expressing radioresistant cancer cell lines. METHODS AND MATERIALS: We examined clonogenic death and the kinetics of apoptotic events in different tumor cell lines overexpressing or underexpressing Hsp27 protein irradiated with photons. The radiosensitive Jurkat cell line, which does not express Hsp27 constitutively or in response to gamma-rays, was stably transfected with Hsp27 complementary DNA. Attenuation of Hsp27 expression was accomplished by antisense or RNAi (interfering RNA) strategies in SQ20B head-and-neck squamous carcinoma, PC3 prostate cancer, and U87 glioblastoma radioresistant cells. RESULTS: We measured concentration-dependent protection against the cytotoxic effects of radiation in Jurkat-Hsp27 cells, which led to a 50% decrease in apoptotic cells at 48 hours in the highest expressing cells. Underlying mechanisms leading to radiation resistance involved a significant increase in glutathione levels associated with detoxification of reactive oxygen species, a delay in mitochondrial collapse, and caspase activation. Conversely, attenuation of Hsp27 in SQ20B cells, characterized by their resistance to apoptosis, sensitizes cells to irradiation. This was emphasized by increased apoptosis, decreased glutathione basal level, and clonogenic cell death. Sensitization to irradiation was confirmed in PC3 and U87 radioresistant cells. CONCLUSION: Hsp27 gene therapy offers a potential adjuvant to radiation-based therapy of resistant tumors.


Asunto(s)
Apoptosis/efectos de la radiación , Silenciador del Gen/fisiología , Proteínas de Choque Térmico/fisiología , Proteínas de Neoplasias/fisiología , Tolerancia a Radiación/fisiología , Apoptosis/fisiología , Caspasas/metabolismo , Línea Celular Tumoral/efectos de la radiación , Regulación hacia Abajo , Activación Enzimática/efectos de la radiación , Rayos gamma , Glioblastoma/metabolismo , Glioblastoma/radioterapia , Glutatión/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/radioterapia , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Células Jurkat/efectos de la radiación , Masculino , Mitocondrias/fisiología , Mitocondrias/efectos de la radiación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Estrés Oxidativo/fisiología , Estrés Oxidativo/efectos de la radiación , Fotones , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/radioterapia , ARN sin Sentido/uso terapéutico , ARN Interferente Pequeño/uso terapéutico , Tolerancia a Radiación/genética , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Transfección/métodos , Ensayo de Tumor de Célula Madre
8.
Cell Stress Chaperones ; 13(3): 313-26, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18330721

RESUMEN

In leukemia cells, hyperthermia enhances tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis. The phenomenon is caspase-dependent and results in membrane changes leading to an increased recognition of TRAIL death receptors by TRAIL. Because either caspase-2 or an apical proteolytic event has been recently proposed to act as an initiator of the cell death mechanism induced by heat shock, we have investigated the hierarchy of caspase activation in cells exposed to the combined heat shock plus TRAIL treatment. We report here that caspases-2, -3, and -8 were the first caspases to be activated. As expected, caspase-8 is required and indispensable during the initiation of this death signaling. Caspase-2 may also participate in the phenomenon but, in contrast to caspase-8, its presence appears dispensable because its depletion by small interfering RNA is devoid of effects. Our observations also suggest a role of caspase-3 and of a particular cleaved form of this caspase during the early signals of heat shock plus TRAIL-induced apoptosis.


Asunto(s)
Apoptosis/fisiología , Caspasas/metabolismo , Fiebre , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Inhibidores de Caspasas , Caspasas/genética , Activación Enzimática , Humanos , Células Jurkat , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/genética
9.
Methods Mol Biol ; 1709: 163-178, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29177658

RESUMEN

Human HspB1 (also denoted as Hsp27) belongs to the family of small (or stress) proteins (sHsps). The family, which contains ten members including αA,B-crystallin polypeptides, is characterized by a conserved C-terminal α-crystallin domain and molecular weights ranging from 20 to 40 kDa. Here, procedures are described for analyzing the dynamic oligomerization and phosphorylation patterns of HspB1 in cells exposed to different environments. Changes in the structural organization of HspB1 can reprogram its interaction with specific partner/client polypeptides. Methods are presented to analyze these interactions using tissue culture cells genetically modified to express different levels of this protein. In addition, the laboratory approaches presented here could be used to test the nine other human sHsp members as well as sHsps from other species.


Asunto(s)
Proteínas de Choque Térmico HSP27/metabolismo , Inmunoprecipitación/métodos , Biología Molecular/métodos , Multimerización de Proteína , Procesamiento Proteico-Postraduccional , Proteínas de Choque Térmico HSP27/análisis , Células HeLa , Proteínas de Choque Térmico , Humanos , Chaperonas Moleculares , Fosforilación , Proteómica/métodos
10.
FEBS Lett ; 581(19): 3665-74, 2007 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-17467701

RESUMEN

Hsp27 and alphaB-crystallin are molecular chaperones that are constitutively expressed in several mammalian cells, particularly in pathological conditions. These proteins share functions as diverse as protection against toxicity mediated by aberrantly folded proteins or oxidative-inflammation conditions. In addition, these proteins share anti-apoptotic properties and are tumorigenic when expressed in cancer cells. This review summarizes the current knowledge about Hsp27 and alphaB-crystallin and the implications, either positive or deleterious, of these proteins in pathologies such as neurodegenerative diseases, myopathies, asthma, cataracts and cancers. Approaches towards therapeutic strategies aimed at modulating the expression and/or the activities of Hsp27 and alphaB-crystallin are presented.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Inflamación/tratamiento farmacológico , Chaperonas Moleculares/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Cadena B de alfa-Cristalina/metabolismo , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/antagonistas & inhibidores , Proteínas de Choque Térmico/química , Humanos , Inflamación/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/efectos de los fármacos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Neoplasias/metabolismo , Conformación Proteica , Cadena B de alfa-Cristalina/antagonistas & inhibidores , Cadena B de alfa-Cristalina/química
11.
Cancer Lett ; 250(1): 117-27, 2007 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-17141951

RESUMEN

We recently reported that, in cultured leukemic T lymphocytes and promyelocytic cells, a mild heat shock treatment (1 h at 42 degrees C) induced a long lasting stimulation of the apoptosis induced by TNF-related apoptosis inducing ligand (TRAIL). On the opposite, no effects were recorded toward normal human T lymphocytes. The apoptogenic efficiency of TRAIL in leukemic lymphocytes is linked to the long lasting increased ability of TRAIL to recognize and bind DR4 and DR5 receptors during hyperthermia. Here, we have analyzed whether this new apoptotic co-treatment could be relevant toward primary cells from patients suffering of chronic lymphocytic leukemia. Analysis of samples from 24 patients with different ages, sex and disease stages revealed that half of them had lymphocytes that, once isolated and analyzed in vitro, positively responded (increase of cell death) to the heat shock plus TRAIL co-treatment. Analysis of the level of expression of various anti-apoptotic proteins in the cell samples revealed a great heterogeneity between patients and no clear relationships could be drawn. Nevertheless, most cell samples that were sensitive to TRAIL plus heat shock induced apoptosis displayed a higher level of cell surface DR4 and DR5 receptors than the non-sensitive counterparts. Hence, analysis of the level of TRAIL surface receptors is a prerequisite for future clinical applications based on this protocol.


Asunto(s)
Calor , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Anciano , Anciano de 80 o más Años , Apoptosis , Membrana Celular/metabolismo , Femenino , Humanos , Hipertermia Inducida , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Masculino , Persona de Mediana Edad , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteínas Recombinantes/farmacología
12.
Mol Cell Biol ; 22(3): 816-34, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11784858

RESUMEN

We previously showed that Hsp27 protects against apoptosis through its interaction with cytosolic cytochrome c. We have revisited this protective activity in murine cell lines expressing different levels of Hsp27. We report that Hsp27 also interferes, in a manner dependent on level of expression, with the release of cytochrome c from mitochondria. Moreover, a decreased level of endogenous Hsp27, which sensitized HeLa cells to apoptosis, reduced the delay required for cytochrome c release and procaspase 3 activation. The molecular mechanism regulating this function of Hsp27 is unknown. In our cell systems, Hsp27 is mainly cytosolic and only a small fraction of this protein colocalized with mitochondria. Moreover, we show that only a very small fraction of cytochrome c interacts with Hsp27, hence excluding a role of this interaction in the retention of cytochrome c in mitochondria. We also report that Bid intracellular relocalization was altered by changes in Hsp27 level of expression, suggesting that Hsp27 interferes with apoptotic signals upstream of mitochondria. We therefore investigated if the ability of Hsp27 to act as an expression-dependent modulator of F-actin microfilaments integrity was linked to the retention of cytochrome c in mitochondria. We show here that the F-actin depolymerizing agent cytochalasin D rapidly induced the release of cytochrome c from mitochondria and caspase activation. This phenomenon was delayed in cells pretreated with the F-actin stabilizer phalloidin and in cells expressing a high level of Hsp27. This suggests the existence of an apoptotic signaling pathway linking cytoskeleton damages to mitochondria. This pathway, which induces Bid intracellular redistribution, is negatively regulated by the ability of Hsp27 to protect F-actin network integrity. However, this upstream pathway is probably not the only one to be regulated by Hsp27 since, in staurosporine-treated cells, phalloidin only partially inhibited cytochrome c release and caspase activation. Moreover, in etoposide-treated cells, Hsp27 still delayed the release of cytochrome c from mitochondria and Bid intracellular redistribution in conditions where F-actin was not altered.


Asunto(s)
Grupo Citocromo c/metabolismo , Proteínas de Choque Térmico , Proteínas de Neoplasias/metabolismo , Células 3T3 , Actinas/metabolismo , Animales , Apoptosis/fisiología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3 , Proteínas Portadoras/metabolismo , Caspasa 3 , Caspasas/metabolismo , Línea Celular , Activación Enzimática , Etopósido/farmacología , Expresión Génica , Glutatión/metabolismo , Proteínas de Choque Térmico HSP27 , Células HeLa , Humanos , Cinética , Ratones , Mitocondrias/metabolismo , Modelos Biológicos , Chaperonas Moleculares , Proteínas de Neoplasias/genética , Estaurosporina/farmacología
13.
Adv Exp Med Biol ; 594: 14-26, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17205671

RESUMEN

Cells possess effective mechanisms to cope with chronic or acute disturbance of homeostasis. Key roles in maintaining or restoring homeostasis are played by the various heat shock or stress proteins (Hsps). Among the Hsps, the group of proteins characterized by low molecular masses (between 20 to 30 kDa) and homology to alpha-crystallin are called small stress proteins (denoted sHsps). The present chapter summarizes the actual knowledge of the protective mechanisms generated by the expression of mammalian Hsp27 (also denoted HspB1 in human) against the cytotoxicity induced by heat shock and oxidative stress. It also describes the anti-apoptotic properties of Hsp27 and their putative consequences in different pathological conditions.


Asunto(s)
Apoptosis , Proteínas de Choque Térmico/metabolismo , Pliegue de Proteína , Animales , Proteínas de Choque Térmico/química , Respuesta al Choque Térmico , Humanos , Mitocondrias/metabolismo , Oxidación-Reducción
14.
Cell Stress Chaperones ; 22(4): 517-529, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28144778

RESUMEN

Constitutively expressed small heat shock protein HspB1 regulates many fundamental cellular processes and plays major roles in many human pathological diseases. In that regard, this chaperone has a huge number of apparently unrelated functions that appear linked to its ability to recognize many client polypeptides that are subsequently modified in their activity and/or half-life. A major parameter to understand how HspB1 is dedicated to interact with particular clients in defined cellular conditions relates to its complex oligomerization and phosphorylation properties. Indeed, HspB1 structural organization displays dynamic and complex rearrangements in response to changes in the cellular environment or when the cell physiology is modified. These structural modifications probably reflect the formation of structural platforms aimed at recognizing specific client polypeptides. Here, I have reviewed data from the literature and re-analyzed my own studies to describe and discuss these fascinating changes in HspB1 structural organization.


Asunto(s)
Proteínas de Choque Térmico HSP27/química , Proteínas de Choque Térmico HSP27/metabolismo , Animales , Puntos de Control del Ciclo Celular , Diferenciación Celular , Proliferación Celular , Proteínas de Choque Térmico , Respuesta al Choque Térmico , Humanos , Chaperonas Moleculares , Neoplasias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Estrés Oxidativo , Fosforilación , Conformación Proteica , Mapas de Interacción de Proteínas , Multimerización de Proteína
15.
FEBS J ; 273(23): 5428-41, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17116244

RESUMEN

Recently, we reported that the transient expression of huntingtin exon1 polypeptide containing polyglutamine tracts of various sizes (httEx1-polyQ) in cell models of Huntington disease generated an oxidative stress whose intensity was CAG repeat expansion-dependent. Here, we have analyzed the intracellular localization of the oxidative events generated by the httEx1-polyQ polypeptides. Analysis of live COS-7 cells as well as neuronal SK-N-SH and PC12 cells incubated with hydroethidine or dichlorofluorescein diacetate revealed oxidation of these probes at the level of the inclusion bodies formed by httEx1-polyQ polypeptides. The intensity and frequency of the oxidative events among the inclusions were CAG repeat expansion-dependent. Electron microscopic analysis of cell sections revealed the presence of oxidation-dependent morphologic alterations in the vicinity of httEx1-polyQ inclusion bodies. Moreover, a high level of oxidized proteins was recovered in partially purified inclusions. We also report that the iron chelator deferroxamine altered the structure, localization and oxidative potential of httEx1-polyQ inclusion bodies. Hence, despite the fact that the formation of inclusion bodies may represent a defense reaction of the cell to eliminate httEx1 mutant polypeptide, this phenomenon appears inherent to the generation of iron-dependent oxidative events that can be deleterious to the cell.


Asunto(s)
Enfermedad de Huntington/metabolismo , Cuerpos de Inclusión/metabolismo , Hierro/metabolismo , Acetilcisteína/farmacología , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Deferoxamina/metabolismo , Exones , Proteínas de Choque Térmico/metabolismo , Cuerpos de Inclusión/ultraestructura , Microscopía Confocal , Oxidación-Reducción , Fenantridinas/metabolismo , Ratas , Proteínas Recombinantes de Fusión
16.
FEBS J ; 273(13): 3076-93, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16817855

RESUMEN

We recently reported that the transient expression of polyglutamine tracts of various size in exon 1 of the huntingtin polypeptide (httEx1) generated abnormally high levels of intracellular reactive oxygen species that directly contributed to cell death. Here, we compared the protection generated by heat shock proteins to that provided by the antioxidant agent N-acetyl-L-cysteine. In cells expressing httEx1 with 72 glutamine repeats (httEx1-72Q), the overexpression of Hsp27 or Hsp70 plus Hdj-1(Hsp40) or treatment of the cells with N-acetyl-L-cysteine inhibited not only mitochondrial membrane potential disruption but also the increase in reactive oxygen species, nitric oxide and protein oxidation. However, only heat shock proteins and not N-acetyl-L-cysteine reduced the size of the inclusion bodies formed by httEx1-72Q. In cells expressing httEx1 polypeptide with 103 glutamine repeats (httEx1-103Q), heat shock proteins neither decreased oxidative damage nor reduced the size of the inclusions. In contrast, N-acetyl-L-cysteine still efficiently decreased the oxidative damage induced by httEx1-103Q polypeptide without altering the inclusions. N-Acetyl-L-cysteine was inactive with regard to proteasome inhibition, whereas heat shock proteins partially restored the caspase-like activity of this protease. These observations suggest some relationships between the presence of inclusion bodies and the oxidative damage induced by httEx1-polyQ.


Asunto(s)
Antioxidantes/farmacología , Proteínas de Choque Térmico/química , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Estrés Oxidativo , Acetilcisteína/química , Animales , Células COS , Chlorocebus aethiops , Exones , Glutamina/química , Proteínas de Choque Térmico HSP27 , Proteínas del Choque Térmico HSP40/química , Proteína Huntingtina , Chaperonas Moleculares , Proteínas de Neoplasias/química , Óxido Nítrico/química , Óxido Nítrico/metabolismo , Péptido Hidrolasas/química , Péptidos/química , Especies Reactivas de Oxígeno
17.
Mol Biol Cell ; 27(11): 1712-27, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27075172

RESUMEN

During cell life, proteins often misfold, depending on particular mutations or environmental changes, which may lead to protein aggregates that are toxic for the cell. Such protein aggregates are the root cause of numerous diseases called "protein conformational diseases," such as myofibrillar myopathy and familial amyotrophic lateral sclerosis. To fight against aggregates, cells are equipped with protein quality control mechanisms. Here we report that NFκB transcription factor is activated by misincorporation of amino acid analogues into proteins, inhibition of proteasomal activity, expression of the R120G mutated form of HspB5 (associated with myofibrillar myopathy), or expression of the G985R and G93A mutated forms of superoxide dismutase 1 (linked to familial amyotrophic lateral sclerosis). This noncanonical stimulation of NFκB triggers the up-regulation of BAG3 and HspB8 expression, two activators of selective autophagy, which relocalize to protein aggregates. Then NFκB-dependent autophagy allows the clearance of protein aggregates. Thus NFκB appears as a central and major regulator of protein aggregate clearance by modulating autophagic activity. In this context, the pharmacological stimulation of this quality control pathway might represent a valuable strategy for therapies against protein conformational diseases.


Asunto(s)
Autofagia/fisiología , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Células HeLa , Humanos , Neuronas Motoras/metabolismo , Conformación Proteica , Pliegue de Proteína , Proteínas Serina-Treonina Quinasas/metabolismo , Estrés Fisiológico/genética , Estrés Fisiológico/fisiología , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Activación Transcripcional , Regulación hacia Arriba , Cadena B de alfa-Cristalina/genética , Cadena B de alfa-Cristalina/metabolismo
18.
Oncotarget ; 7(40): 65808-65824, 2016 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-27588467

RESUMEN

Our recent study has shown that αA-crystallin appears to act as a tumor suppressor in pancreas. Here, we analyzed expression patterns of αA-crystallin in the pancreatic tumor tissue and the neighbor normal tissue from 74 pancreatic cancer patients and also pancreatic cancer cell lines. Immunocytochemistry revealed that αA-crystallin was highly expressed in the normal tissue from 56 patients, but barely detectable in the pancreatic tumor tissue. Moreover, a low level of αA-crystallin predicts poor prognosis for patients with pancreatic duct adenocarcinoma (PDAC). In the 12 pancreatic cell lines analyzed, except for Capan-1 and Miapaca-2 where the level of αA-crystallin was about 80% and 65% of that in the control cell line, HPNE, the remaining pancreatic cancer cells have much lower αA-crystallin levels. Overexpression of αA-crystallin in MiaPaca-1 cells lacking endogenous αA-crystallin significantly decreased its tumorigenicity ability as shown in the colony formation and wound healing assays. In contrast, knockdown of αA-crystallin in the Capan-1 cells significantly increased its tumorigenicity ability as demonstrated in the above assays. Together, our results further demonstrate that αA-crystallin negatively regulates pancreatic tumorigenesis and appears to be a prognosis biomarker for PDAC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Ductal Pancreático/patología , Transformación Celular Neoplásica/patología , Neoplasias Pancreáticas/patología , Cadena A de alfa-Cristalina/metabolismo , Anciano , Apoptosis , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/cirugía , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Masculino , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/cirugía , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas
19.
Antioxid Redox Signal ; 7(3-4): 436-45, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15706091

RESUMEN

Murine small stress protein [heat shock protein 25 (Hsp25)] expression confers thermotolerance and protection against oxidative stress. Hsp25 is an oligomeric ATP-independent phospho-chaperone that can generate a glutathione-dependent pro-reducing state in cells that are normally devoid of small stress protein constitutive expression. Hsp25 contains only one cysteine residue (position 141) that is highly susceptible to oxidation. We have explored the significance of this reactive residue by generating a mutant in which cysteine-141 was substituted by an alanine residue (C141A mutant). We report here that the C141A mutant did not form dimers when expressed in either murine L929 or human HeLa cells, hence, demonstrating that cysteine-141 regulates Hsp25 dimer formation. The C141A mutant also interfered with the dimerization of human Hsp27, a constitutively expressed small stress protein in HeLa cells. The mutated polypeptide showed a decreased ability to multimerize, but its expression was still able to induce cellular protection against oxidative stress. The C141A mutant was, however, less efficient than the wild-type protein in counteracting staurosporine-induced apoptosis, and it showed no in vivo chaperone activity. Hence, the cellular protection mediated against different stressors may require specific structural organizations of Hsp25 that are differently altered by the mutation. Of interest, when expressed concomitantly with wild-type Hsp25, the C141A polypeptide induced a dominant-negative effect, a phenomenon that may result from the ability of small stress proteins to interact and multimerize with each other.


Asunto(s)
Cisteína/genética , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Estrés Oxidativo , Sustitución de Aminoácidos , Animales , Apoptosis , Línea Celular , Dimerización , Células HeLa , Proteínas de Choque Térmico/química , Humanos , Ratones , Chaperonas Moleculares/metabolismo , Mutación , Proteínas de Neoplasias/química
20.
Antioxid Redox Signal ; 7(3-4): 414-22, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15706088

RESUMEN

Small stress proteins [small heat shock proteins (sHsps)] are molecular chaperones that modulate the ability of cells to respond to oxidative stress. The current knowledge concerning the protective mechanism generated by the expression of mammalian heat shock protein-27 (Hsp27) that allows cells to increase their resistance to oxidative stress is presented. We describe the effects mediated by Hsp27 expression toward crucial enzymes such as glucose-6-phosphate dehydrogenase and glutathione reductase that uphold glutathione in its reduced form. New data are presented showing that the expression of sHsps correlates with a drastic decrease in the intracellular level of iron, a catalyzer of hydroxyl radical (OH( . )) generation. A decreased ability of sHsps expressing cells to concentrate iron will therefore end up in a decreased level of oxidized proteins. In addition, we propose a role of Hsp27 in the presentation of oxidized proteins to the proteasome degradation machinery. We also present an analysis of several Hsp27 mutants that suggests that the C-terminal part of this stress protein is essential for its protective activity against oxidative stress.


Asunto(s)
Glutatión/metabolismo , Proteínas de Choque Térmico/fisiología , Hierro/metabolismo , Estrés Oxidativo , Animales , Regulación hacia Abajo , Proteínas de Choque Térmico/genética , Homeostasis , Ratones , Oxidación-Reducción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA