Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Cancer Sci ; 114(1): 152-163, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36102493

RESUMEN

Recent comprehensive analyses of mtDNA and orthogonal RNA-sequencing data revealed that in numerous human cancers, mtDNA copy numbers and mtRNA amounts are significantly reduced, followed by low respiratory gene expression. Under such conditions (called mt-Low), cells encounter severe cell proliferation defects; therefore, they must acquire countermeasures against this fatal disadvantage during malignant transformation. This study elucidated a countermeasure against the mt-Low condition-induced antiproliferative effects in hepatocellular carcinoma (HCC) cells. The mechanism relied on the architectural transcriptional regulator HMGA2, which was preferably expressed in HCC cells of the mt-Low type in vitro and in vivo. Detailed in vitro analyses suggest that HMGA2 regulates insulin-like growth factor binding protein 1 (IGFBP1) expression, leading to AKT activation, which then phosphorylates the cyclin-dependent kinase inhibitor (CKI), P27KIP1, and facilitates its ubiquitin-mediated degradation. Accordingly, intervention in the HMGA2 function by RNAi resulted in an increase in P27KIP1 levels and an induction of senescence-like cell proliferation inhibition in mt-Low-type HCC cells. Conclusively, the HMGA2/IGFBP1/AKT axis has emerged as a countermeasure against P27KIP1 CKI upregulation under mt-Low conditions, thereby circumventing cell proliferation inhibition and supporting the tumorigenic state. Notably, similar to in vitro cell lines, HMGA2 was likely to regulate IGFBP1 expression in HCC in vivo, thereby contributing to poor patient prognosis. Considering the significant number of cases under mt-Low or the threat of CKI upregulation cancer-wide, the axis is noteworthy as a vulnerability of cancer cells or target for tumor-agnostic therapy inducing irreversible cell proliferation inhibition via CKI upregulation in a large population with cancer.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , ARN , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias Hepáticas/patología , ADN Mitocondrial , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina , Proliferación Celular/genética , Inhibidores de Proteínas Quinasas/farmacología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica
2.
Biochem Biophys Res Commun ; 649: 1-9, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36738577

RESUMEN

Aggressive cancers, such as triple-negative breast cancer (TNBC), are mostly fatal because of their potential to metastasize to distant organs. Cancer cells acquire various abilities to metastasize, including resistance to anoikis, an apoptotic cell death induced by loss of anchorage to the extracellular matrix. Transcriptional coactivator with PDZ binding motif (TAZ) and Yes-associated protein (YAP), the downstream effectors of the Hippo pathway, regulate cell- and tissue-level architectures by responding to mechanical microenvironments of cells, including the cell-extracellular matrix interaction. The Hippo pathway is frequently disrupted in cancer cells, and TAZ and YAP are irrelevantly activated, potentially resulting in anchorage-independent survival/proliferation of cancer cells and metastatic progression. The study aims to investigate the roles of TAZ and YAP in anoikis resistance in basal-like (BL) TNBC cells, which comprise a major subtype (>70%) of TNBC. We found that TAZ and YAP had nonredundant roles in anchorage-independent cancer cell survival or anoikis resistance. Particularly, TAZ was indispensable for anoikis resistance in BL-TNBC cells but not for survival of non-transformed mammary epithelial cells (MECs). In contrast, YAP, a paralog of TAZ, was indispensable for survival of both non-transformed MECs and cancer cells. Therefore, TAZ might be a preferable therapeutic target against dissemination of aggressive cancer cells without killing normal cells. Interestingly, TAZ was abnormally stabilized in BL-TNBC cells under non-adherent conditions, which promoted anoikis resistance. Furthermore, OTUB1, a deubiquitinating enzyme, was responsible for the stabilization of TAZ in detached BL-TNBC cells. Importantly, simultaneous high expression of TAZ and OTUB1 was associated with poor prognosis in BC. Thus, OTUB1 has emerged as a potentially druggable target. Successful inhibition of OTUB1 enzymatic activity is expected to downregulate TAZ and eventually prevents metastasis of aggressive cancers, such as BL-TNBC.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Neoplasias de la Mama Triple Negativas , Humanos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Anoicis/fisiología , Neoplasias de la Mama Triple Negativas/patología , Proteínas Señalizadoras YAP , Enzimas Desubicuitinizantes/metabolismo , Microambiente Tumoral
3.
Cancer Sci ; 112(8): 3205-3217, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34036687

RESUMEN

Previously, we reported that non-apoptotic cell death was induced in non-malignant mammary epithelial cells (HMECs) upon loss of anchorage during 48 h incubation in suspension. In this study, we examined HMECs in suspension at an earlier time point and found that most of them lost attachment ability to substrata when replated, although >80% were alive. This suggested that HMECs lost reattachment ability (RA) prior to cell death upon detachment. Concomitant with the loss of RA, a decrease in the levels of ß1 and ß4 integrin was observed. In sharp contrast, breast cancer cells retained integrin levels, reattached to substrata, and formed colonies after exposure to anchorage loss as efficiently as those maintained under adherent conditions. Such RA of cancer cells is essential for the metastatic process, especially for establishing adhesion contact with ECM in the secondary organ after systemic circulation. Further analysis suggested that sustained levels of ß4 integrin, which was mediated by Rac1, was critical for RA after anchorage loss and lung metastasis of breast cancer cells. In the cancer cells, persistent Rac1 activity enhanced escape of ß4 integrin from lysosomal degradation depending on actin-related protein 2/3 and TBC1D2, a GTPase-activating protein of Rab7 GTPase. Notably, simultaneous high expression of ITGB4 and RAC1 was associated with poor prognosis in patients with breast cancer. Therefore, ß4 integrin and Rac1 are attractive therapeutic targets to eliminate RA in cancer cells, thereby preventing the initial step of colonization at the secondary organ during metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Integrina beta4/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Proteína de Unión al GTP rac1/metabolismo , Adhesión Celular , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Lisosomas/metabolismo , Células MCF-7 , Pronóstico
4.
Exp Cell Res ; 389(1): 111889, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32032602

RESUMEN

The copy number of mitochondrial DNA (mtDNA) is decreased in most cancer types, including hepatocellular carcinoma (HCC), compared to normal counterparts. However, a decrease in mtDNA usually leads to defects in cell proliferation, which contradicts the robustness of cancer cell proliferation. In this study, we found that four out of seven HCC cell lines were of the mtDNA-less type. Interestingly, FOXM1, a member of the FOX transcription factor family, was highly expressed in a subset of them with proliferative potential maintained. B-MYB, a partner of FOXM1, was also expressed in the same cell lines. RNAi-mediated experiments demonstrated that when FOXM1/B-MYB was silenced in the cell lines, cell cycle-related genes were downregulated, while p21Cip1 was induced with senescence-associated ß-galactosidase, resulting in G1/S cell cycle arrest. These results suggest that high expression of FOXM1/B-MYB is critical for sustaining cell proliferation in mtDNA-less cells. In addition, we found that high expression of FOXM1 was mediated by the deubiquitinating enzyme, OTUB1, in one cell line. Thus, interference with FOXM1/B-MYB expression, such as through OTUB1 inhibition, may induce a dormant state of senescence-like proliferation arrest in mtDNA-less cancer cells. This finding may be utilized for the development of precision medicine for relevant cancers.


Asunto(s)
Carcinoma Hepatocelular/genética , Proliferación Celular/genética , ADN Mitocondrial/genética , Proteína Forkhead Box M1/genética , Neoplasias Hepáticas/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Células Cultivadas , Variaciones en el Número de Copia de ADN , Regulación Neoplásica de la Expresión Génica , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Neoplasias Hepáticas/patología , Regulación hacia Arriba/genética
5.
Bioorg Med Chem Lett ; 29(18): 2659-2663, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31371134

RESUMEN

Reactive oxygen species (ROS) are known to be produced during the amyloid beta (Aß) aggregation process. Both ROS production and Aß fibril formation can result in nerve cell injury. Proanthocyanidins are oligomers of catechin that can act as inhibitors of Aß aggregation. Procyanidin B3 (Cat-Cat), the dimer of (+)-catechin, can easily cross the blood-brain barrier. Previously, we synthesized two derivatives of Cat-Cat, namely Cat-PCat and PCat-PCat, in which the geometry of one or both catechin molecules in Cat-Cat was constrained to be planar. The antioxidative activities of Cat-PCat and PCat-PCat were found to be stronger than that of Cat-Cat, with PCat-PC at exhibiting the most potent activity. These compounds are predicted to protect against Aß-induced neurotoxicity via inhibition of Aß aggregation as well as by antioxidative effects toward Aß-induced intracellular ROS generation. PCat-PCat exhibited the most potent neuroprotective effects against Aß-induced cytotoxicity, which resulted from inhibition of ß-sheet structure formation during the Aß aggregation process. PCat-PCat may be a promising lead compound for the treatment of Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Antioxidantes/farmacología , Biflavonoides/farmacología , Catequina/farmacología , Fármacos Neuroprotectores/farmacología , Proantocianidinas/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Antioxidantes/síntesis química , Antioxidantes/química , Biflavonoides/síntesis química , Biflavonoides/química , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Catequina/síntesis química , Catequina/química , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/química , Proantocianidinas/síntesis química , Proantocianidinas/química , Agregado de Proteínas/efectos de los fármacos , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Relación Estructura-Actividad
6.
Cancer Sci ; 107(7): 963-71, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27094710

RESUMEN

Mitochondria are multifunctional organelles; they have been implicated in various aspects of tumorigenesis. In this study, we investigated a novel role of the basal electron transport chain (ETC) activity in cell proliferation by inhibiting mitochondrial replication and transcription (mtR/T) using pharmacological and genetic interventions, which depleted mitochondrial DNA/RNA, thereby inducing ETC deficiency. Interestingly, mtR/T inhibition did not decrease ATP levels despite deficiency in ETC activity in different cell types, including MDA-MB-231 breast cancer cells, but it severely impeded cell cycle progression, specifically progression during G2 and/or M phases in the cancer cells. Under these conditions, the expression of a group of cell cycle regulators was downregulated without affecting the growth signaling pathway. Further analysis suggested that the transcriptional network organized by E2F1 was significantly affected because of the downregulation of E2F1 in response to ETC deficiency, which eventually resulted in the suppression of cell proliferation. Thus, in this study, the E2F1-mediated ETC-dependent mechanism has emerged as the regulatory mechanism of cell cycle progression. In addition to E2F1, FOXM1 and BMYB were also downregulated, which contributed specifically to the defects in G2 and/or M phase progression. Thus, ETC-deficient cancer cells lost their growing ability, including their tumorigenic potential in vivo. ETC deficiency abolished the production of reactive oxygen species (ROS) from the mitochondria and a mitochondria-targeted antioxidant mimicked the deficiency, thereby suggesting that ETC activity signaled through ROS production. In conclusion, this novel coupling between ETC activity and cell cycle progression may be an important mechanism for coordinating cell proliferation and metabolism.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Factor de Transcripción E2F1/metabolismo , Redes Reguladoras de Genes , Neoplasias de la Mama/genética , Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , División Celular , Línea Celular Tumoral , Proliferación Celular , Inhibición de Contacto , Proteínas de Unión al ADN/deficiencia , Regulación hacia Abajo , Transporte de Electrón/genética , Proteína Forkhead Box M1/metabolismo , Fase G2 , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Mitocondriales/deficiencia , Fenotipo , Especies Reactivas de Oxígeno/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/deficiencia
7.
Bioorg Med Chem Lett ; 26(22): 5468-5471, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27789140

RESUMEN

Amyloid-ß (Aß) deposition and oxidative stress observed in the brains of patients with Alzheimer's disease (AD) are important targets for therapeutic intervention. In this study, we conjugated the antioxidants caffeic acid (CA) and dihydrocaffeic acid (DHCA) to Aß1-42 C-terminal motifs (Aßx-42: x=38, 40) to synthesize CA-Aßx-42 and DHCA-Aßx-42, respectively. Among the compounds, CA-Aß38-42 exhibited potent inhibitory activity against Aß1-42 aggregation and scavenged Aß1-42-induced intracellular oxidative stress. Moreover, CA-Aß38-42 significantly protected human neuroblastoma SH-SY5Y cells against Aß1-42-induced cytotoxicity, with an IC50 of 4µM. These results suggest that CA-Aß38-42 might be a potential lead for the treatment of AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/farmacología , Amiloide/antagonistas & inhibidores , Antioxidantes/farmacología , Ácidos Cafeicos/farmacología , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Agregado de Proteínas/efectos de los fármacos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Amiloide/metabolismo , Péptidos beta-Amiloides/química , Antioxidantes/química , Ácidos Cafeicos/química , Línea Celular Tumoral , Humanos , Fármacos Neuroprotectores/química , Estrés Oxidativo/efectos de los fármacos , Fragmentos de Péptidos/química
8.
Bioorg Med Chem ; 24(18): 4138-4143, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27407032

RESUMEN

Two hallmarks of Alzheimer's disease (AD) observed in the brains of patients with the disease include oxidative injury and deposition of protein aggregates comprised of amyloid-ß (Aß) variants. To inhibit these toxic processes, we synthesized antioxidant-conjugated peptides comprised of Trolox and various C-terminal motifs of Aß variants, TxAßx-n (x=34, 36, 38, 40; n=40, 42, 43). Most of these compounds were found to exhibit anti-aggregation activities. Among them, TxAß36-42 significantly inhibited Aß1-42 aggregation, showed potent antioxidant activity, and protected SH-SY5Y cells from Aß1-42-induced cytotoxicity. Thus, this method represents a promising strategy for developing multifunctional AD therapeutic agents.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/farmacología , Antioxidantes/farmacología , Cromanos/farmacología , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Agregado de Proteínas/efectos de los fármacos , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Antioxidantes/química , Cromanos/química , Diseño de Fármacos , Humanos , Fragmentos de Péptidos/química , Especies Reactivas de Oxígeno/metabolismo
9.
Biochem Biophys Res Commun ; 443(3): 821-7, 2014 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-24342608

RESUMEN

Transforming growth factor (TGF)-ß is a pro-oncogenic cytokine that induces the epithelial-mesenchymal transition (EMT), a crucial event in tumor progression. During TGF-ß-mediated EMT in NMuMG mouse mammary epithelial cells, we observed sustained increases in reactive oxygen species (ROS) levels in the cytoplasm and mitochondria with a concomitant decrease in mitochondrial membrane potential and intracellular glutathione levels. In pseudo ρ0 cells, whose respiratory chain function was impaired, the increase in intracellular ROS levels was abrogated, suggesting an important role of mitochondrial activity as a trigger for TGF-ß-stimulated ROS generation. In line with this, TGF-ß-mediated expression of the EMT marker fibronectin was inhibited not only by chemicals that interfere with ROS signaling but also by exogenously expressed mitochondrial thioredoxin (TXN2) independent of Smad signaling. Of note, TGF-ß-mediated induction of HMGA2, a central mediator of EMT and metastatic progression, was similarly impaired by TXN2 expression, revealing a novel mechanism involving a thiol oxidation reaction in mitochondria, which regulates TGF-ß-mediated gene expression associated with EMT.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Regulación de la Expresión Génica/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Tiorredoxinas/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Animales , Línea Celular , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Células HEK293 , Proteínas HMGA/metabolismo , Humanos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Glándulas Mamarias Animales/citología , Ratones , Mitocondrias/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
10.
J Biol Chem ; 287(46): 38854-65, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-23007394

RESUMEN

Anchorage loss elicits a set of responses in cells, such as transcriptional changes, in order to prevent inappropriate cell growth in ectopic environments. However, the mechanisms underlying these responses are poorly understood. In this study, we investigated the transcriptional up-regulation of cyclin-dependent kinase inhibitor p21(Cip1) during anchorage loss, which is important for cell cycle arrest of nonadherent cells in the G1 phase. Up-regulation was mediated by an upstream element, designated as the detachment-responsive element (DRE), that contained Kruppel-like factor 4 (KLF4) and runt-related transcription factor 1 (RUNX1) recognition sites; both of these together were necessary for transactivation, as individually they were insufficient. RNAi experiments revealed that KLF4 and a multidomain adaptor protein, hydrogen peroxide-inducible clone 5 (HIC-5), were critically involved in DRE transactivation. The role of HIC-5 in this mechanism was to tether KLF4 to DNA sites in response to cellular detachment. In addition, further analysis suggested that oligomerization and subsequent nuclear matrix localization of HIC-5, which was accelerated spontaneously in cells during anchorage loss, was assumed to potentiate the scaffolding function of HIC-5 in the nucleus and consequently regulate p21(Cip1) transcription in a manner responding to anchorage loss. At the RUNX1 site, a LIM-only protein, CRP2, imposed negative regulation on transcription, which appeared to be removed by anchorage loss and contributed to increased transcriptional activity of DRE together with regulation at the KLF4 sites. In conclusion, this study revealed a novel transcriptional mechanism that regulated gene expression in a detachment-dependent manner, thereby contributing to anchorage-dependent cell growth.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas con Dominio LIM/metabolismo , Activación Transcripcional , Animales , Sitios de Unión , Adhesión Celular , ADN/química , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Factor 4 Similar a Kruppel , Ratones , Ratones Endogámicos C3H , Modelos Biológicos , Unión Proteica , Fracciones Subcelulares
11.
Cancer Sci ; 103(10): 1803-10, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22726539

RESUMEN

In most human cancers, somatic mutations have been identified in the mtDNA; however, their significance remains unclear. We recently discovered that NMuMG mouse mammary epithelial cells, when deprived of mitochondria or following inhibition of respiratory activity, undergo epithelial morphological disruption accompanied with irregular edging of E-cadherin, the appearance of actin stress fibers, and an altered gene expression profile. In this study, using the mtDNA-less pseudo ρ0 cells obtained from NMuMG mouse mammary epithelial cells, we examined the roles of two mitochondrial stress-associated transcription factors, cAMP-responsive element-binding protein (CREB) and C/EBP homologous protein-10 (CHOP), in the disorganization of epithelial phenotypes. We found that the expression of matrix metalloproteinase-13 and that of GADD45A, SNAIL and integrin α1 in the ρ0 cells were regulated by CHOP and CREB, respectively. Of note, knockdown and pharmacological inhibition of CREB ameliorated the disrupted epithelial morphology. It is interesting to note that the expression of high mobility group AT-hook 2 (HMGA2), a non-histone chromatin protein implicated in malignant neoplasms, was increased at the protein level through the CREB pathway. Here, we reveal how the activation of the CREB/HMGA2 pathway is implicated in the repression of integrin α1 expression in HepG2 human cancer cells, highlighting the importance of the CREB/HMGA2 pathway in malignant transformation associated with mitochondrial dysfunction, thereby raising the possibility that the pathway indirectly interferes with the cell-cell adhesion structure by influencing the cell-extracellular matrix adhesion status. Overall, the data suggest that mitochondrial dysfunction potentially contributes to neoplastic transformation of epithelial cells through the activation of these transcriptional pathways.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Mitocondrias/patología , Animales , Western Blotting , Línea Celular Tumoral , ADN Mitocondrial/metabolismo , Proteína HMGA2/metabolismo , Humanos , Ratones , Mitocondrias/metabolismo , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/fisiología
12.
J Mol Cell Cardiol ; 50(1): 77-86, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20933520

RESUMEN

Forces associated with blood flow are crucial not only for blood vessel development but also for regulation of vascular pathology. Although there have been many studies characterizing the responses to mechanical stimuli, molecular mechanisms linking biological responses to mechanical forces remain unclear. Hic-5 (hydrogen peroxide-inducible clone-5) is a focal adhesion adaptor protein proposed as a candidate for a mediator of mechanotransduction. In the present study, we generated Hic-5-deficient mice by targeted mutation. Mice lacking Hic-5 are viable and fertile, and show no obvious histological abnormalities including vasculature. However, after wire injury of the femoral artery in Hic-5 deficient mice, histological recovery of arterial media was delayed due to enhanced apoptosis of vascular wall cells, whereas neointima formation was enhanced. Stretch-induced apoptosis was enhanced in cultured vascular smooth muscle cells (vascular SMCs) from Hic-5 deficient mice. Mechanical stress also induced the alteration of intracellular distribution of vinculin from focal adhesions to the whole cytoplasm in SMCs. Immunoelectron microscopic study of vascular SMCs from a wire-injured artery demonstrated that vinculin was dispersed in the nucleus and the cytoplasm in Hic-5-deficient mice whereas vinculin was localized mainly in the sub-plasma membrane region in wild type mice. Our findings indicate that Hic-5 may serve as a key regulator in mechanosensitive vascular remodeling.


Asunto(s)
Apoptosis/fisiología , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Animales , Apoptosis/genética , Southern Blotting , Western Blotting , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/genética , Adhesiones Focales/genética , Adhesiones Focales/metabolismo , Proteínas con Dominio LIM , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Microscopía Inmunoelectrónica , Modelos Biológicos , Miocitos del Músculo Liso/ultraestructura , Especies Reactivas de Oxígeno/metabolismo , Vinculina/metabolismo
13.
ACS Omega ; 5(36): 23164-23174, 2020 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-32954167

RESUMEN

Silibinin (Sib), one of the main components of milk thistle extract, has attracted considerable attention because of its various biological activities, which include antioxidant activity and potential effects in diabetes and Alzheimer's disease (AD). In a previous study, we synthesized catechin analogues by constraining the geometries of (+)-catechin and (-)-epicatechin. The constrained analogues exhibited enhanced bioactivities, with the only major difference between the two being their three-dimensional structures. The constrained geometry in (+)-catechin resulted in a high degree of planarity (PCat), while (-)-epicatechin failed to maintain planarity (PEC). The three-dimensional structure of Sib may be related to its ability to inhibit aggregation of amyloid beta (Aß). We therefore introduced PCat and PEC into Sib to demonstrate how the constrained molecular geometry and differences in three-dimensional structures may enhance such activities. Introduction of PCat into Sib (SibC) resulted in effective inhibition of Aß aggregation, α-glucosidase activity, and cell growth, suggesting that not only reduced flexibility but also the high degree of planarity may enhance the biological activity. SibC is expected to be a promising lead compound for the treatment of several diseases.

14.
FASEB J ; 22(2): 428-36, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17848623

RESUMEN

Recent evidence suggests that bone marrow-derived cells may contribute to repair and lesion formation following vascular injury. In most studies, bone marrow-derived cells were tracked by transplanting exogenous cells into bone marrow that had been compromised by irradiation. It remains to be determined whether endogenous circulating progenitors actually contribute to arterial remodeling under physiological conditions. Here, we established a parabiotic model in which two mice were conjoined subcutaneously without any vascular anastomosis. When wild-type mice were joined with transgenic mice that expressed green fluorescent protein (GFP) in all tissues, GFP-positive cells were detected not only in the peripheral blood but also in the bone marrow of the wild-type mice. The femoral arteries of the wild-type mice were mechanically injured by insertion of a large wire. At 4 wk, there was neointima hyperplasia that mainly consisted of alpha-smooth muscle actin-positive cells. GFP-positive cells were readily detected in the neointima (14.8+/-4.5%) and media (31.1+/-8.8%) of the injured artery. Some GFP-positive cells expressed alpha-smooth muscle actin or an endothelial cell marker. These results indicate that circulating progenitors contribute to re-endothelialization and neointimal formation after mechanical vascular injury even in nonirradiated mice.


Asunto(s)
Movimiento Celular , Células Madre/citología , Actinas/metabolismo , Animales , Trasplante de Médula Ósea , Genes Reporteros/genética , Hiperplasia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Inmunoelectrónica , Parabiosis , Células Madre/metabolismo , Células Madre/efectos de la radiación
15.
Exp Cell Res ; 314(10): 2131-40, 2008 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-18433744

RESUMEN

We found that a specific isoform of hepatocyte nuclear factor 4alpha (HNF-4alpha), HNF-4alpha8, was expressed in mouse mammary epithelial NMuMG cells, and that its expression was repressed by TGF-beta. The repression was interfered by dominant negative forms of activin receptor-like kinase 5 (ALK5) and Smad3, and sensitive to cycloheximide, suggesting the involvement of additional protein(s) as well as ALK5 and Smad3 in the repression. Further study showed that high mobility group A2 (HMGA2), which is reported to be directly upregulated by Smads, repressed HNF-4alpha8 expression. Therefore, it is likely that HMGA2 mediates the downregulation of HNF-4alpha8 downstream of ALK5 and Smads To determine the significance of the downregulation of HNF-4alpha8 in TGF-beta signaling, we performed DNA microarray analysis and extracted a subgroup of TGF-beta1-regulated genes, including tenascin C and tissue inhibitor of metalloproteinase 3 (TIMP-3), whose regulation by TGF-beta1 was attenuated by forced expression of HNF-4alpha8. HMGA2 has recently emerged as a transcriptional organizer of TGF-beta signaling, regulating several key factors involved in epithelial-mesenchymal transition (EMT). In this study, we identified an isoform of HNF-4alpha as a new target downstream of HMGA2 and assigned a new role to HNF-4alpha in the TGF-beta signaling/transcriptional cascade driven by ALK5/Smad/HMGA2 and associated with the malignant transformation of cells.


Asunto(s)
Células Epiteliales/fisiología , Regulación de la Expresión Génica , Factor Nuclear 4 del Hepatocito/metabolismo , Glándulas Mamarias Animales/citología , Isoformas de Proteínas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Transformación Celular Neoplásica , Células Cultivadas , Regulación hacia Abajo , Células Epiteliales/citología , Femenino , Perfilación de la Expresión Génica , Proteína HMGA2/genética , Proteína HMGA2/metabolismo , Factor Nuclear 4 del Hepatocito/genética , Humanos , Ratones , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Isoformas de Proteínas/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Retroviridae/genética , Retroviridae/metabolismo , Transducción de Señal/fisiología , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/genética
16.
Curr Drug Metab ; 20(5): 361-376, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30947665

RESUMEN

BACKGROUND: Evidence has revealed that renal impairment can affect the systemic exposure of drugs which are predominantly eliminated via the liver. The modulation of drug-metabolizing enzymes and transporters expressed in the liver and/or small intestine by diverse entities, including uremic toxins, in systemic circulation of patients with severe renal failure is considered as the cause of atypical pharmacokinetics, which sometimes induce undesirable adverse events that are especially critical for drugs with narrow therapeutic window such as anticancer drugs. A dosing strategy for anticancer drugs in these patients needs to be established. METHODS: The effects of renal impairment on the systemic exposure and safety of anticancer drugs were summarized. The proposed mechanisms for the alterations in the pharmacokinetics of these anticancer drugs were also discussed. RESULTS: Changes in pharmacokinetics and clinical response were reported in 9 out of 10 cytotoxic anticancer drugs investigated, although available information was limited and sometimes controversial. Systemic exposure of 3 out of 16 tyrosine kinase inhibitors was higher in patients with severe renal failure than that in patients with normal kidney function. An increase in systemic exposure of anticancer drugs in patients with renal impairment is likely to be observed for substrates of OATP1B1, despite the limited evidence. CONCLUSION: The molecular basis for the effect of uremia on non-renal drug elimination still needed to be clarified with further studies to generate generalizable concepts, which may provide insights into establishing better clinical usage of anticancer drugs, i.e. identifying patients at risk and dose adjustment.


Asunto(s)
Antineoplásicos/farmacocinética , Hígado/metabolismo , Inhibidores de Proteínas Quinasas/farmacocinética , Insuficiencia Renal/metabolismo , Animales , Humanos
17.
FEBS J ; 286(3): 459-478, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30281903

RESUMEN

Matrix metalloproteinases (MMPs) are tissue-remodeling enzymes involved in the processing of various biological molecules. MMPs also play important roles in cancer metastasis, contributing to angiogenesis, intravasation of tumor cells, and cell migration and invasion. Accordingly, unraveling the signaling pathways controlling MMP activities could shed additional light on cancer biology. Here, we report a molecular axis, comprising the molecular adaptor hydrogen peroxide-inducible clone-5 (HIC-5), NADPH oxidase 4 (NOX4), and mitochondria-associated reactive oxygen species (mtROS), that regulates MMP9 expression and may be a target to suppress cancer metastasis. We found that this axis primarily downregulates mtROS levels which stabilize MMP9 mRNA. Specifically, HIC-5 suppressed the expression of NOX4, the source of the mtROS, thereby decreasing mtROS levels and, consequently, destabilizing MMP9 mRNA. Interestingly, among six cancer cell lines, only EJ-1 and MDA-MB-231 cells exhibited upregulation of NOX4 and MMP9 expression after shRNA-mediated HIC-5 knockdown. In these two cell lines, activating RAS mutations commonly occur, suggesting that the HIC-5-mediated suppression of NOX4 depends on RAS signaling, a hypothesis that was supported experimentally by the introduction of activated RAS into mammary epithelial cells. Notably, HIC-5 knockdown promoted lung metastasis of MDA-MB-231 cancer cells in mice. The tumor growth of HIC-5-silenced MDA-MB-231 cells at the primary sites was comparable to that of control cells. Consistently, the invasive properties of the cells, but not their proliferation, were enhanced by the HIC-5 knockdown in vitro. We conclude that NOX4-mediated mtROS signaling increases MMP9 mRNA stability and affects cancer invasiveness but not tumor growth.


Asunto(s)
Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas con Dominio LIM/genética , Neoplasias Pulmonares/genética , Mitocondrias/metabolismo , NADPH Oxidasa 4/genética , Especies Reactivas de Oxígeno/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Senescencia Celular , Células Epiteliales/metabolismo , Células Epiteliales/patología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Adhesiones Focales/metabolismo , Adhesiones Focales/patología , Xenoinjertos , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/antagonistas & inhibidores , Proteínas con Dominio LIM/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos NOD , Mitocondrias/patología , NADPH Oxidasa 4/metabolismo , Invasividad Neoplásica , Estrés Oxidativo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal
18.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(11): 1606-1618, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31376475

RESUMEN

Long-chain acyl-coenzyme A synthetases (ACSLs) are a family of enzymes that convert free long-chain fatty acids into their acyl-coenzyme A (CoA) forms. ACSL4, belonging to the ACSL family, shows a preferential use of arachidonic acid (AA) as its substrate and plays a role in the remodeling of AA-containing phospholipids by incorporating free AA. However, little is known about the roles of ACSL4 in inflammatory responses. Here, we assessed the roles of ACSL4 on the effector functions of bone marrow-derived macrophages (BMDMs) obtained from mice lacking ACSL4. Liquid chromatography-tandem mass spectrometry analysis revealed that various highly unsaturated fatty acid (HUFA)-derived fatty acyl-CoA species were markedly decreased in the BMDMs obtained from ACSL4-deficient mice compared with those in the BMDMs obtained from wild-type mice. BMDMs from ACSL4-deficient mice also showed a reduced incorporation of HUFA into phosphatidylcholines. The stimulation of BMDMs with lipopolysaccharide (LPS) elicited the release of prostaglandins (PGs), such as PGE2, PGD2 and PGF2α, and the production of these mediators was significantly enhanced by ACSL4 deficiency. In contrast, neither the LPS-induced release of cytokines, such as IL-6 and IL-10, nor the endocytosis of zymosan or dextran was affected by ACSL4 deficiency. These results suggest that ACSL4 has a crucial role in the maintenance of HUFA composition of certain phospholipid species and in the incorporation of free AA into the phospholipids in LPS-stimulated macrophages. ACSL4 dysfunction may facilitate inflammatory responses by an enhanced eicosanoid storm.


Asunto(s)
Coenzima A Ligasas/metabolismo , Ácidos Grasos Insaturados/metabolismo , Macrófagos/metabolismo , Fosfolípidos/metabolismo , Animales , Ácido Araquidónico/metabolismo , Células Cultivadas , Coenzima A Ligasas/genética , Femenino , Ratones , Ratones Noqueados , Especificidad por Sustrato
19.
Cancer Chemother Pharmacol ; 83(6): 1127-1135, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30972456

RESUMEN

PURPOSE: Several retrospective studies have shown that the antitumor efficacy of capecitabine-containing chemotherapy decreases when co-administered with a proton pump inhibitor (PPI). Although a reduction in capecitabine absorption by PPIs was proposed as the underlying mechanism, the effects of PPIs on capecitabine pharmacokinetics remain unclear. We prospectively examined the effects of rabeprazole on the pharmacokinetics of capecitabine and its metabolites. METHODS: We enrolled patients administered adjuvant capecitabine plus oxaliplatin (CapeOX) for postoperative colorectal cancer (CRC) patients and metastatic CRC patients receiving CapeOX with/without bevacizumab. Patients receiving a PPI before registration were allocated to the rabeprazole group, and the PPI was changed to rabeprazole (20 mg/day) at least 1 week before the initiation of capecitabine treatment. On day 1, oral capecitabine (1000 mg/m2) was administered 1 h after rabeprazole intake. Oxaliplatin (and bevacizumab) administration on day 1 was shifted to day 2 for pharmacokinetic analysis of the first capecitabine dose. Plasma concentrations of capecitabine, 5'-deoxy-5-fluorocytidine, 5'-deoxy-5-fluorouridine, and 5-fluorouracil were analyzed by high-performance liquid chromatography. Effects of rabeprazole on inhibition of cell proliferation by each capecitabine metabolite were examined with colon cancer cells (COLO205 and HCT116). RESULTS: Five and 9 patients enrolled between September 2017 and July 2018 were allocated to rabeprazole and control groups, respectively. No significant effects of rabeprazole on area under the plasma concentration-time curve divided by capecitabine dose for capecitabine and its three metabolites were observed. Rabeprazole did not affect the proliferation inhibition of colon cancer cells by the respective capecitabine metabolites. CONCLUSION: Rabeprazole does not affect capecitabine pharmacokinetics.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Capecitabina/administración & dosificación , Neoplasias Colorrectales/tratamiento farmacológico , Inhibidores de la Bomba de Protones/administración & dosificación , Rabeprazol/administración & dosificación , Anciano , Antimetabolitos Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica , Área Bajo la Curva , Capecitabina/farmacocinética , Proliferación Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacocinética , Interacciones Farmacológicas , Femenino , Floxuridina/farmacocinética , Fluorouracilo/farmacocinética , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Inhibidores de la Bomba de Protones/farmacología , Rabeprazol/farmacología
20.
Biochem Biophys Res Commun ; 376(4): 682-7, 2008 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-18812162

RESUMEN

Focal adhesion components are targets for biochemical and mechanical stimuli that evoke crucial injury. Hic-5 (hydrogen peroxide-inducible clone 5) is a multidomain adaptor protein which is implicated in the regulation of integrin signaling in focal adhesion. The aim of this research was to test the hypothesis that Hic-5, a focal adhesion LIM protein expressed in smooth muscle cells, is involved in dynamic processes by pathological stimuli in the vessel wall. Here, we describe the analysis of the function of Hic-5 using a mouse model of vascular injury that may mimic balloon angioplasty. At 4 days after vascular injury, marked down-regulation of the Hic-5 expression was observed in the smooth muscle layer, and local delivery of the Hic-5 using adenovirus vectors repressed injury-induced neointimal expansion. In addition, Hic-5 reduced cells migration into three-dimensional collagen gels, and the forced expression of Hic-5 in cells embedded in the collagen gel matrix repressed the expression of uPA that participates in smooth muscle cell migration. These results suggest that Hic-5 modulates cellular responses to pathological stimuli in the vessel wall.


Asunto(s)
Angioplastia de Balón/efectos adversos , Traumatismos de las Arterias Carótidas/metabolismo , Proteínas del Citoesqueleto/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Adenoviridae , Animales , Traumatismos de las Arterias Carótidas/patología , Movimiento Celular , Proliferación Celular , Colágeno/química , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Adhesiones Focales , Geles/química , Humanos , Hiperplasia , Proteínas con Dominio LIM , Ratones , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Ratas , Transducción Genética , Túnica Íntima/lesiones , Túnica Íntima/metabolismo , Túnica Íntima/patología , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA