Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Transplantation ; 108(3): e36-e48, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38126420

RESUMEN

BACKGROUND: Although heart transplantation is the definitive treatment for heart failure in eligible patients, both acute and chronic transplant rejection frequently occur. Protein phosphatase 2A (PP2A) activity is critical in maintaining tissue and organ homeostasis. In this study, we evaluated the effect of a novel class of small molecule activators of PP2A (SMAPs) on allograft rejection in a mouse heterotopic heart transplantation model. METHODS: Recipient mice were administered with DT-061 (a pharmaceutically optimized SMAP) or vehicle by oral gavage beginning 1 d after transplantation. Histological and immunofluorescence analyses were performed to examine allograft rejection. Regulatory T cells (Treg) from recipient spleens were subjected to flow cytometry and RNA sequencing analysis. Finally, the effect of DT-061 on smooth muscle cells (SMCs) migration and proliferation was assessed. RESULTS: DT-061 treatment prolonged cardiac allograft survival. SMAPs effectively suppressed the inflammatory immune response while increasing Treg population in the allografts, findings corroborated by functional analysis of RNA sequencing data derived from Treg of treated splenic tissues. Importantly, SMAPs extended immunosuppressive agent cytotoxic T lymphocyte-associated antigen-4-Ig-induced cardiac transplantation tolerance and allograft survival. SMAPs also strongly mitigated cardiac allograft vasculopathy as evidenced by a marked reduction of neointimal hyperplasia and SMC proliferation. Finally, our in vitro studies implicate suppression of MEK/ERK pathways as a unifying mechanism for the effect of PP2A modulation in Treg and SMCs. CONCLUSIONS: PP2A activation prevents cardiac rejection and prolongs allograft survival in a murine model. Our findings highlight the potential of PP2A activation in improving alloengraftment in heart transplantation.


Asunto(s)
Rechazo de Injerto , Trasplante de Corazón , Humanos , Ratones , Animales , Proteína Fosfatasa 2/farmacología , Supervivencia de Injerto , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trasplante de Corazón/efectos adversos
2.
Stem Cell Rev Rep ; 19(6): 1981-1993, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37243830

RESUMEN

Osteogeinc differentiation from mesenchymal stem cells (MSCs) into osteoblasts is a key step for bone tissue engineering in regenerative medicine. The insight into regulatory mechanism of osteogenesis of MSCs facilitates achieving better recovery effect. Long non-coding RNAs are regarded as a family of important moderators in osteogenesis. In this study, we found a novel lncRNA, lnc-PPP2R1B was up-regulated during osteogenesis of MSCs by Illumina HiSeq transcritome sequencing. We demonstrated lnc-PPP2R1B overexpression promoted osteogenesis and knockdown of lnc-PPP2R1B inhibited osteogenesis of MSCs. Mechanically, it physically interacted with and up-regulated heterogeneous nuclear ribonucleoprotein L Like (HNRNPLL), which is a master regulator of activation-induced alternative splicing in T cells. We found lnc-PPP2R1B knockdown or HNRNPLL knockdown decreased transcript-201 of Protein Phosphatase 2A, Regulatory Subunit A, Beta Isoform (PPP2R1B) while increased transcript-203 of PPP2R1B, and did not affect transcript-202/204/206. PPP2R1B is a constant regulatory subunit of protein phosphatase 2 (PP2A), which activates Wnt/ß-catenin pathway by removing phosphorylation and stabilization of ß-catenin and translocation into nucleus. The transcript-201 retained exon 2 and 3, compared to transcript-203. And it was reported the exon 2 and 3 of PPP2R1B were one part of B subunit binding domain on A subunit in PP2A trimer, and therefore retaining exon 2 and 3 promised formation and enzyme function of PP2A. Finally, lnc-PPP2R1B promoted ectopic osteogenesis in vivo. Conclusively, lnc-PPP2R1B mediated alternative splicing of PPP2R1B through retaining exon 2 and 3 by interacting with HNRNPLL and then promoted osteogenesis, which may facilitate an in-depth understanding of function and mechanism of lncRNAs in osteogenesis. Lnc-PPP2R1B interacted with HNRNPLL, and regulated alternative splicing of PPP2R1B through retaining exon 2 and 3, which preserved enzyme function of PP2A and enhanced dephosphorylation and nuclear translocation of ß-catenin, thereby promoting Runx2 and OSX expression and then osteogenesis. And it provided experimental data and potential target for promoting bone formation and bone regeneration.


Asunto(s)
Ribonucleoproteína Heterogénea-Nuclear Grupo L , Células Madre Mesenquimatosas , Empalme Alternativo/genética , beta Catenina/genética , beta Catenina/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo L/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo L/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo L/farmacología , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2/farmacología , Humanos
3.
J Pediatr Surg ; 58(6): 1145-1154, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36907775

RESUMEN

BACKGROUND: The tumor suppressor, protein phosphatase 2A (PP2A), is downregulated in hepatoblastoma. We aimed to examine the effects of two novel compounds of the tricyclic sulfonamide class, ATUX-3364 (3364) and ATUX-8385 (8385), designed to activate PP2A without causing immunosuppression, on human hepatoblastoma. METHODS: An established human hepatoblastoma cell line, HuH6, and a human hepatoblastoma patient-derived xenograft, COA67, were treated with increasing doses of 3364 or 8385, and viability, proliferation, cell cycle and motility were investigated. Cancer cell stemness was evaluated by real-time PCR and tumorsphere forming ability. Effects on tumor growth were examined using a murine model. RESULTS: Treatment with 3364 or 8385 significantly decreased viability, proliferation, cell cycle progression and motility in HuH6 and COA67 cells. Both compounds significantly decreased stemness as demonstrated by decreased abundance of OCT4, NANOG, and SOX2 mRNA. The ability of COA67 to form tumorspheres, another sign of cancer cell stemness, was significantly diminished by 3364 and 8385. Treatment with 3364 resulted in decreased tumor growth in vivo. CONCLUSION: Novel PP2A activators, 3364 and 8385, decreased hepatoblastoma proliferation, viability, and cancer cell stemness in vitro. Animals treated with 3364 had decreased tumor growth. These data provide evidence for further investigation of PP2A activating compounds as hepatoblastoma therapeutics.


Asunto(s)
Hepatoblastoma , Neoplasias Hepáticas , Humanos , Animales , Ratones , Hepatoblastoma/tratamiento farmacológico , Hepatoblastoma/genética , Hepatoblastoma/metabolismo , Neoplasias Hepáticas/genética , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2/farmacología , Proteína Fosfatasa 2/uso terapéutico , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Línea Celular Tumoral , Proliferación Celular
4.
Biomed Pharmacother ; 160: 114350, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36804120

RESUMEN

Long-term use of low-toxic natural products holds the promise for eradicating cancer stem cells. In this study, we report that luteolin, a natural flavonoid, attenuates the stemness of ovarian cancer stem cells (OCSCs) by directly binding to KDM4C and epigenetic suppression of PPP2CA/YAP axis. Ovarian cancer stem like cells (OCSLCs) isolated by suspension culture and CD133 + ALDH+ cell sorting was employed as OCSCs model. The maximal non-toxic dose of luteolin suppressed stemness properties, including sphere-forming capacity, the expression of OCSCs markers, sphere-initiating and tumor-initiating capacities, as well as the percentage of CD133 + ALDH+ cells of OCSLCs. Mechanistic study showed that luteolin directly binds to KDM4C, blocks KDM4C-induced histone demethylation of PPP2CA promoter, inhibits PPP2CA transcription and PPP2CA-mediated YAP dephosphorylation, thereby attenuating YAP activity and the stemness of OCSLCs. Furthermore, luteolin sensitized OCSLCs to traditional chemotherapeutic drugs in vitro and in vivo. In summary, our work revealed the direct target of luteolin and the underlying mechanism of the inhibitory effect of luteolin on the stemness of OCSCs. This finding thus suggests a novel therapeutic strategy for eradicating human OCSCs driven by KDM4C.


Asunto(s)
Luteolina , Neoplasias Ováricas , Femenino , Humanos , Línea Celular Tumoral , Epigénesis Genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Luteolina/farmacología , Luteolina/uso terapéutico , Células Madre Neoplásicas/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2/farmacología , Proteína Fosfatasa 2/uso terapéutico , Proteínas Señalizadoras YAP/metabolismo
5.
Inflamm Res ; 72(3): 373-385, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36562794

RESUMEN

OBJECTIVE: Pleckstrin homology domain and leucine-rich repeat protein phosphatase 2 (PHLPP2) is linked to various pathological states. However, whether PHLPP2 mediates diabetic retinopathy is unaddressed. This work explored the biological function of PHLPP2 in modulating high glucose (HG)-elicited damage of retinal ganglion cells (RGCs), an in vitro model for studying diabetic retinopathy. METHODS: Mouse RGCs were treated with HG to establish a cell model. PHLPP2 was silenced by transfecting specific shRNAs targeting PHLPP2. RT-qPCR, immunoblotting, CCK-8 assay, flow cytometry, TUNEL assay, and ELISA were carried out. RESULTS: Significant increases in PHLPP2 levels were observed in cultured RGCs exposed to HG. The severe damages evoked by HG to RGCs were remarkably weakened in PHLPP2-silenced RGCs, including improved cell survival, attenuated cell apoptosis, repressed oxidative stress, and prohibited proinflammatory response. The silencing of PHLPP2 strengthened the activation of Nrf2 in HG-treated RGCs via modulation of the Akt-GSK-3ß axis. Interruption of the Akt-GSK-3ß axis reversed PHLPP2-silencing-elicited Nrf2 activation. The protective effects of PHLPP2 silencing on HG-induced injury of RGCs were diminished by Nrf2 inhibition. CONCLUSIONS: The loss of PHLPP2 was beneficial for HG-injured RGCs through the effect on the Akt-GSK-3ß-Nrf2 pathway. This work suggests a possible role of PHLPP2 in diabetic retinopathy.


Asunto(s)
Retinopatía Diabética , Proteínas Proto-Oncogénicas c-akt , Ratones , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2/farmacología , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Transducción de Señal , Proteínas Repetidas Ricas en Leucina , Dominios Homólogos a Pleckstrina , Células Ganglionares de la Retina/metabolismo , Retinopatía Diabética/genética , Estrés Oxidativo , Glucosa/farmacología , Apoptosis
6.
J Affect Disord ; 317: 265-277, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36031001

RESUMEN

BACKGROUND: The putative mechanisms underlying the efficacy of the US Food and Drug Administration-approved antipsychotic drug paliperidone for the treatment of schizophrenia deserve additional investigation, which is the aim of the present animal study. METHODS: The behavioral activities of mice were recorded in the open field test and light-dark box test. The effects of paliperidone on MK-801-induced neuronal damage in the prefrontal cortex were tested by flow cytometry, TUNEL staining assays, and ROS staining assays. The neuroprotective effects of paliperidone on neural dendrites and synapses were evaluated using Golgi staining and Sholl analysis. An adenovirus vector containing a Ca2+ indicator was used to monitor the calcium ion concentration in the prefrontal cortex. The expression levels of protein phosphatase 2A (PP2A) and phosphatase and tensin homolog (PTEN) were investigated using Western blotting. RESULTS: The data showed that MK-801 caused stereotyped behavior in mice and induced synaptic damage and dendritic spine impairment compared with the control, whereas paliperidone ameliorated these changes. Moreover, paliperidone reversed MK-801-induced decreases in PP2A and PTEN levels in prefrontal cortical neurons. Furthermore, in primary cultured cortical neurons and HT-22 cells, paliperidone inhibited cell apoptosis caused by MK-801. In particular, pretreatment with the PP2A inhibitor LB-100 significantly restrained the protective effects of paliperidone on MK-801-treated neurons and on locomotor activity and stereotypical behavior of mice. LIMITATIONS: Whether other proteins are involved in this pathway and how the pathway works have not been revealed. CONCLUSION: Our data show that paliperidone alleviates neuronal damage induced by MK-801 via the PP2A/PTEN pathway.


Asunto(s)
Antipsicóticos , Fármacos Neuroprotectores , Animales , Antipsicóticos/farmacología , Calcio/metabolismo , Maleato de Dizocilpina/farmacología , Ratones , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Fosfohidrolasa PTEN/metabolismo , Palmitato de Paliperidona/metabolismo , Palmitato de Paliperidona/farmacología , Corteza Prefrontal/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2/farmacología , Especies Reactivas de Oxígeno/metabolismo , Tensinas/metabolismo
7.
Phytomedicine ; 101: 154116, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35525235

RESUMEN

BACKGROUND: Despite recent advances in the treatment of squamous cell skin cancer (SCSC), the disease persists, and treatment resistance develops. Thus, identifying new targets and developing new therapeutic approaches showing low vulnerability to drug resistance is highly needed. PURPOSE: This study aimed to reveal a novel targeted phytotherapeutic strategy for SCSC treatment alone or in combination with standard targeted anticancer molecules. STUDY DESIGN: A library of natural products was utilized to identify molecules that inhibit the growth of skin cancer cells. The anticancer potential of the selected compound was evaluated in human skin squamous carcinoma models, in vitro and in vivo. A comprehensive ingenuity pathway analysis (IPA) strategy and molecular biology technology was adopted to investigate the therapeutic mechanisms in human SCSC. METHODS: The Matrigel invasion chamber, foci formation and soft agar colony formation assays were employed to study the cells invasion and migration potential in vitro. In vivo antitumor effects were evaluated in DMBA/TPA-induced skin papilloma and A431 human skin squamous carcinoma xenograft tumor models. An integrative IPA was employed to identify mechanisms and protein targets in human SCSC.Compounds synergies were determined by the bliss model and evaluated using human SCSC cell lines and xenograft tumors. Histological staining, immunofluorescence imaging, real-time PCR, Western blots, and flow cytometric analyses were employed to analyze apoptosis and cell signaling mechanisms. RESULTS: We identified (+)-cyanidan-3-ol (CD-3) as a selective compound for inhibiting the growth of SCSC cell lines. CD-3 inhibited tumor growth and burden without apparent toxicity and prolonged the survival of tumor-bearing mice. CD-3 inhibitory effects on SCSC growth are mediated via cell cycle arrest and caspase-dependent apoptosis induction. Mechanistic studies showed that CD-3 activates PP2A via inhibiting CIP2A and produces tumor growth inhibitory effects via promoting dephosphorylation of oncogenic AKT/mTOR signaling proteins in SCSC cells and xenograft tumors in a PP2A dependent manner. Furthermore, the combination of CD-3 and mTOR inhibitors (mTORi) synergistically reduced oncogenic phenotypes. CONCLUSIONS: Our study suggests that PP2A activation is an effective strategy for SCSC treatment and the CD-3 and mTORi combination may serve as a promising treatment for SCSC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias Cutáneas , Animales , Humanos , Ratones , Apoptosis , Autoantígenos/genética , Autoantígenos/metabolismo , Autoantígenos/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Línea Celular Tumoral , Células Epiteliales/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Neoplasias Cutáneas/tratamiento farmacológico , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Exp Dermatol ; 31(8): 1154-1164, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35298048

RESUMEN

Psoriasis, a common skin disease, endangers human physiological and mental health; however, its pathogenesis remains unclear. Keratinocyte proliferation is a typical pathological characteristic of psoriasis. Serine/threonine protein phosphatase 2A (PP2A) is one of the most important phosphatases for maintaining normal phosphorylation levels in humans. PP2Acα is the alpha subtype of the PP2A C subunit (encoded by PPP2CA), which maintains the catalytic functions of PP2A. Epidermal growth factor receptor (EGFR) is activated by phosphorylation (p-EGFR) to regulate the downstream signalling pathway to promote epidermal cell proliferation. Previous studies have found that PPP2CA induced epidermal hyperplasia, keratinization and other pathological phenomena similar to those in mouse models of psoriasis. The present study showed that PP2Acα negatively regulated EGFR phosphorylation and epidermal cell proliferation, and EGFR inhibitors could alleviate PP2Acα by inhibiting epidermal cell proliferation. This study further examined the effect of mechanisms on epidermal cell proliferation and the downstream signalling pathway of EGFR using molecular technological methods to explore new ideas for treating psoriasis.


Asunto(s)
Proteína Fosfatasa 2 , Psoriasis , Animales , Proliferación Celular , Receptores ErbB/metabolismo , Humanos , Hiperplasia , Ratones , Fosforilación , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
9.
Mol Pharm ; 19(3): 895-903, 2022 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-35113575

RESUMEN

Chronic lymphocytic leukemia (CLL) is the most common form of leukemia in adults. The disease is characterized by the accumulation of tumoral B cells resulting from a defect of apoptosis. We have in vitro and in vivo preclinically validated a tumor-penetrating peptide (named TT1) coupled to an interfering peptide (IP) that dissociates the interaction between the serine/threonine protein phosphatase 2A (PP2A) from its physiological inhibitor, the oncoprotein SET. This TT1-IP peptide has an antitumoral effect on CLL, as shown by the increased survival of mice bearing xenograft models of CLL, compared to control mice. The peptide did not show toxicity, as indicated by the mouse body weight and the biochemical parameters, such as renal and hepatic enzymes. In addition, the peptide-induced apoptosis in vitro of primary tumoral B cells isolated from CLL patients but not of those isolated from healthy patients. Finally, the peptide had approximately 5 h half-life in human serum and showed pharmacokinetic parameters compatible with clinical development as a therapeutic peptide against CLL.


Asunto(s)
Leucemia Linfocítica Crónica de Células B , Animales , Apoptosis , Linfocitos B/metabolismo , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/metabolismo , Ratones , Péptidos/farmacología , Péptidos/uso terapéutico , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2/farmacología , Proteína Fosfatasa 2/uso terapéutico
10.
JCI Insight ; 52019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31335320

RESUMEN

Chronic inflammation causes target organ damage in patients with systemic autoimmune diseases. The factors that allow this protracted response are poorly understood. We analyzed the transcriptional regulation of PPP2R2B (B55ß), a molecule necessary for the termination of the immune response, in patients with autoimmune diseases. Altered expression of B55ß conditioned resistance to cytokine withdrawal-induced death (CWID) in patients with autoimmune diseases. The impaired upregulation of B55ß was caused by inflammation-driven hypermethylation of specific cytosines located within a regulatory element of PPP2R2B preventing CTCF binding. This phenotype could be induced in healthy T cells by exposure to TNF-α. Our results reveal a gene whose expression is affected by an acquired defect, through an epigenetic mechanism, in the setting of systemic autoimmunity. Because failure to remove activated T cells through CWID could contribute to autoimmune pathology, this mechanism illustrates a vicious cycle through which autoimmune inflammation contributes to its own perpetuation.


Asunto(s)
Apoptosis/efectos de los fármacos , Enfermedades Autoinmunes/metabolismo , Metilación de ADN , Proteínas del Tejido Nervioso/metabolismo , Proteína Fosfatasa 2/metabolismo , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Factor de Unión a CCCTC/metabolismo , Citocinas/metabolismo , Citosina/metabolismo , Metilación de ADN/efectos de los fármacos , Regulación de la Expresión Génica , Humanos , Inflamación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/farmacología , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/farmacología , Linfocitos T , Regulación hacia Arriba
11.
Brain Behav ; 9(3): e01187, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30729695

RESUMEN

OBJECTIVES: We evaluated the protective effects of protein phosphatase 2A (PP2A)/tristetraprolin (TTP) against brain edema in a rat model of cerebral hemorrhage, bleeding in the brain that occurs in tissues and ventricles. TTP is a well-known mRNA-binding protein and essential regulatory molecule for gene expression. METHODS: Cerebral hemorrhage was induced in male albino rats divided into four homogeneous groups: normal control (I), control (II), PP2A siRNA (III), and scrambled siRNA (IV). Neurological scores, caspase-3 mRNA and protein expression, PP2A and TTP protein expression, apoptosis, and water content in the brain were determined. RESULTS: The neurological score decreased substantially to 8.2 in rats in which cerebral hemorrhage was induced and was further reduced to 7.4 and 7.7 in groups III and IV, respectively. Caspase-3 expression increased significantly by 90% in group II and by 26.9% in group III. Apoptosis increased by 26.1% in rats in which cerebral hemorrhage was induced and increased considerably by 35.3% and 33.4% in groups III and IV, respectively. PP2A and TTP protein expression increased significantly by 87% and 59%, as compared to their respective sham controls. However, PP2A and TTP siRNA treatment reduced the protein expression of PP2A and TTP in groups III and IV. The water content in the brain increased significantly by 77.4% in rats in which cerebral hemorrhage was induced (group II), as compared to the sham group. The water content in the brain increased by 84.1% and 78.7% in groups III and IV, respectively. CONCLUSION: Taken together, these data indicate that TTP has a protective role against brain edema by reducing inflammation, apoptosis, and water content in the brain at 48 hr after cerebral hemorrhage. Our findings may be useful for developing important approaches to treating brain injury.


Asunto(s)
Apoptosis , Edema Encefálico , Hemorragia Cerebral , Proteína Fosfatasa 2/farmacología , Tristetraprolina/farmacología , Animales , Encéfalo/metabolismo , Edema Encefálico/etiología , Edema Encefálico/metabolismo , Edema Encefálico/prevención & control , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/tratamiento farmacológico , Hemorragia Cerebral/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Masculino , Fármacos Neuroprotectores/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
12.
Nucleic Acids Res ; 46(15): 7844-7857, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30016500

RESUMEN

The molecular mechanisms that underlie the neurological manifestations of patients with inherited diseases of vitamin B12 (cobalamin) metabolism remain to date obscure. We observed transcriptomic changes of genes involved in RNA metabolism and endoplasmic reticulum stress in a neuronal cell model with impaired cobalamin metabolism. These changes were related to the subcellular mislocalization of several RNA binding proteins, including the ELAVL1/HuR protein implicated in neuronal stress, in this cell model and in patient fibroblasts with inborn errors of cobalamin metabolism and Cd320 knockout mice. The decreased interaction of ELAVL1/HuR with the CRM1/exportin protein of the nuclear pore complex and its subsequent mislocalization resulted from hypomethylation at R-217 produced by decreased S-adenosylmethionine and protein methyl transferase CARM1 and dephosphorylation at S221 by increased protein phosphatase PP2A. The mislocalization of ELAVL1/HuR triggered the decreased expression of SIRT1 deacetylase and genes involved in brain development, neuroplasticity, myelin formation, and brain aging. The mislocalization was reversible upon treatment with siPpp2ca, cobalamin, S-adenosylmethionine, or PP2A inhibitor okadaic acid. In conclusion, our data highlight the key role of the disruption of ELAVL1/HuR nuclear export, with genomic changes consistent with the effects of inborn errors of Cbl metabolisms on brain development, neuroplasticity and myelin formation.


Asunto(s)
Transporte Biológico/genética , Proteína 1 Similar a ELAV/metabolismo , Carioferinas/metabolismo , Enfermedades Metabólicas/genética , Proteínas de Unión al ARN/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Vitamina B 12/metabolismo , Animales , Encéfalo/patología , Proteínas Adaptadoras de Señalización CARD/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Estrés del Retículo Endoplásmico/genética , Humanos , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ácido Ocadaico/farmacología , Fosforilación , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2/farmacología , ARN Mensajero/metabolismo , S-Adenosilmetionina/farmacología , Sirtuina 1/biosíntesis , Proteína Exportina 1
13.
J Biol Chem ; 293(32): 12525-12534, 2018 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-29945972

RESUMEN

Adaptations and responses to stress conditions are fundamental processes that all cells must accomplish to maintain or restore cellular homeostasis. Cells have a plethora of response pathways to mitigate the effect of different environmental stressors. The transcriptional regulators transcription factor EB (TFEB) and transcription factor binding to IGHM enhancer 3 (TFE3) play a key role in the control of these stress pathways. Therefore, understanding their regulation under different stress conditions is of great interest. Here, using a range of human and murine cells, we show that TFEB and TFE3 are activated upon induction of acute oxidative stress by sodium arsenite via an mTOR complex 1 (mTORC1)-independent process. We found that the mechanism of arsenite-stimulated TFEB and TFE3 activation instead involves protein phosphatase 2A (PP2A)-mediated dephosphorylation at Ser-211 and Ser-321, respectively. Depletion of either the catalytic (PPP2CA+B) or regulatory (PPP2R2A/B55α) subunits of PP2A, as well as PP2A inactivation with the specific inhibitor okadaic acid, abolished TFEB and TFE3 activation in response to sodium arsenite. Conversely, PP2A activation by ceramide or the sphingosine-like compound FTY720 was sufficient to induce TFE3 nuclear translocation. MS analysis revealed that PP2A dephosphorylates TFEB at several residues, including Ser-109, Ser-114, Ser-122, and Ser-211, thus facilitating TFEB activation. Overall, this work identifies a critical mechanism that activates TFEB and TFE3 without turning off mTORC1 activity. We propose that this mechanism may enable some cell types such as immune or cancer cells that require simultaneous TFEB/TFE3 and mTORC1 signaling to survive and achieve robust cell growth in stressful environments.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Estrés Oxidativo , Proteína Fosfatasa 2/farmacología , Animales , Arsenitos/farmacología , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Células Cultivadas , Humanos , Ratones , Fosforilación , Transducción de Señal , Compuestos de Sodio/farmacología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
14.
World J Urol ; 34(1): 89-95, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25981400

RESUMEN

INTRODUCTION AND OBJECTIVES: Reactive oxygen species (ROS) are produced during the interaction between oxalate/calcium oxalate monohydrate (COM) crystals and renal epithelial cells and are responsible for the various cellular responses through the activation of NADPH oxidase (Nox). Ox and COM also activate the renin-angiotensin-aldosterone system (RAAS). Aldosterone stimulates ROS production through activation of Nox with the involvement of mineralocorticoid receptor (MR), Rac1 and mitogen-activated protein kinases (MAPK). We investigated RAAS pathways in vivo in an animal model of hyperoxaluria and in vitro by exposing renal epithelial cells to COM crystals. METHODS: Hyperoxaluria was induced in male SD rats by administering ethylene glycol. One group of rats was additionally given spironolactone. Total RNA was extracted and subjected to genomic microarrays to obtain global transcriptome data. Normal rat kidney cell line (NRK-52E) was incubated with aldosterone(10(-7) M) and COM(67 µg/cm(2)) with or without spironolactone(10(-5) M), a selective inhibitor of SRC family of kinases; protein phosphatase 2(pp2) (10(-5) M) and Nox inhibitor; diphenylene iodonium (DPI) (10(-5) M). RESULTS: Relative expression of genes encoding for AGT, angiotensin receptors 1b and 2, Renin 1, Cyp11b, HSD11B2, Nr3c2, NOx4 and Rac1 was upregulated in the kidneys of rats with hyperoxaluria. Treatment with spironolactone reversed the effect of hyperoxaluria. Both aldosterone and COM crystals activated Nox and Rac1 expression in NRK52E, while spironolactone inhibited Nox and Rac1 expression. Increased Rac1 expression was significantly attenuated by treatment with PP2 and spironolactone. CONCLUSIONS: Results indicate that hyperoxaluria-induced production of ROS, injury and inflammation are in part associated with the activation of Nox through renin-angiotensin-aldosterone pathway.


Asunto(s)
Oxalato de Calcio/metabolismo , Hiperoxaluria/genética , NADPH Oxidasas/metabolismo , ARN Mensajero/metabolismo , Sistema Renina-Angiotensina/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/efectos de los fármacos , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Angiotensinógeno/efectos de los fármacos , Angiotensinógeno/genética , Angiotensinógeno/metabolismo , Animales , Línea Celular , Citocromo P-450 CYP11B2/efectos de los fármacos , Citocromo P-450 CYP11B2/genética , Citocromo P-450 CYP11B2/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Glicol de Etileno/toxicidad , Perfilación de la Expresión Génica , Hiperoxaluria/inducido químicamente , Hiperoxaluria/metabolismo , Masculino , Antagonistas de Receptores de Mineralocorticoides/farmacología , NADPH Oxidasa 4 , NADPH Oxidasas/efectos de los fármacos , NADPH Oxidasas/genética , Compuestos Onio/farmacología , Proteína Fosfatasa 2/farmacología , ARN Mensajero/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de Angiotensina/efectos de los fármacos , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Receptores de Mineralocorticoides/efectos de los fármacos , Receptores de Mineralocorticoides/genética , Receptores de Mineralocorticoides/metabolismo , Renina/efectos de los fármacos , Renina/genética , Renina/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Espironolactona/farmacología , Esteroide 11-beta-Hidroxilasa/efectos de los fármacos , Esteroide 11-beta-Hidroxilasa/genética , Esteroide 11-beta-Hidroxilasa/metabolismo , Proteína de Unión al GTP rac1/efectos de los fármacos , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
15.
Free Radic Res ; 49(10): 1210-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25968938

RESUMEN

While ischemic preconditioning (IPC) and other cardioprotective interventions have been proposed to protect the heart from ischemia/reperfusion (I/R) injury by inhibiting mitochondrial complex I activity upon reperfusion, the exact mechanism underlying the modulation of complex I activity remains elusive. This study was aimed to test the hypothesis that IPC modulates complex I activity at reperfusion by activating mitochondrial Src tyrosine kinase, and induces cardioprotection against I/R injury. Isolated rat hearts were preconditioned by three cycles of 5-min ischemia and 5-min reperfusion prior to 30-min index ischemia followed by 2 h of reperfusion. Mitochondrial Src phosphorylation (Tyr(416)) was dramatically decreased during I/R, implying inactivation of Src tyrosine kinase by I/R. IPC increased mitochondrial Src phosphorylation upon reperfusion and this was inhibited by the selective Src tyrosine kinase inhibitor PP2. IPC's anti-infarct effect was inhibited by the selective Src tyrosine kinase inhibitor PP2. Complex I activity was significantly increased upon reperfusion, an effect that was prevented by IPC in a Src tyrosine kinase-dependent manner. In support, Src and phospho-Src were found in complex I. Furthermore, IPC prevented hypoxia/reoxygenation-induced mitochondrial reactive oxygen species (ROS) generation and cellular injury in rat cardiomyocytes, which was revoked by PP2. Finally, IPC reduced LDH release induced by both hypoxia/reoxygenation and simulated ischemia/reperfusion, an effect that was reversed by PP2 and Src siRNA. These data suggest that mitochondrial Src tyrosine kinase accounts for the inhibitory action of IPC on complex I and mitochondrial ROS generation, and thereby plays a role in the cardioprotective effect of IPC.


Asunto(s)
Complejo I de Transporte de Electrón/antagonistas & inhibidores , Precondicionamiento Isquémico Miocárdico , Mitocondrias Cardíacas/enzimología , Isquemia Miocárdica/enzimología , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/prevención & control , Familia-src Quinasas/fisiología , Animales , Línea Celular , Complejo I de Transporte de Electrón/fisiología , Técnicas In Vitro , Masculino , Mioblastos Cardíacos/metabolismo , Infarto del Miocardio/patología , Miocitos Cardíacos/enzimología , Fosforilación , Proteína Fosfatasa 2/farmacología , Procesamiento Proteico-Postraduccional , Interferencia de ARN , ARN Interferente Pequeño/genética , Ratas , Ratas Wistar , Daño por Reperfusión/enzimología , Superóxidos/metabolismo
16.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 36(3): 234-40, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24997813

RESUMEN

OBJECTIVE: To explore and compare the effects of propofol, ginsenoside Rg-1, protein phosphatae-2A, and lithium on the learning and memory and the concentration of glutamic acid in hippocampus after the electroconvulsive therapy (ECT) in the model of depressed rats induced after the removal of olfactory bulb. METHODS: The depressed rats were randomized into ECT intervention (two levels:no disposition and a course of electroconvulsive shock) and drug intervention (five levels:microinjection of saline injection, propofol, ginsenoside Rg-1, protein phosphatae-2A, and lithium, 20 g/L). Learning and memory were evaluated using the Morris water maze test within 24 h after the course of ECT. Glutamate contents in the hippocampus of rats were examined using high-performance liquid chromatography. RESULTS: Both propofol alone and ECT alone induced the impairment of learning and memory in depressed rats, but their combination alleviated the such impairment caused by ECT. Ginsenoside Rg-1, protein phosphatae-2A ,and lithium had no obvious effect on the leaning and improved the learning and memory when in combination with ECT. There was a synergic effect between ECT intervention and drug intervention. ECT remarkably increased the glutamate content in the hippocampus of depressed rats, which could be reduced by both propofol and ginsenoside Rg-1. Protein phosphatae-2A and lithium did not affect glutamate content in the hippocampus of depressed rats before and after ECT. CONCLUSIONS: ECT can increase the content of glutamate in hippocampus and thus cause the impairment of learning and memory in depressed rats. Propofol and ginsenoside Rg-1 can ameliorate the impairment by reducing the content of glutamate in hippocampus. Protein phosphatae-2A and lithium may also improve the learning and memory in depressed rats.


Asunto(s)
Electrochoque , Ginsenósidos/farmacología , Litio/farmacología , Propofol/farmacología , Proteína Fosfatasa 2/farmacología , Animales , Ácido Glutámico/metabolismo , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
17.
ChemMedChem ; 3(12): 1878-92, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19025735

RESUMEN

Cantharidin (1) and its derivatives are of significant interest as serine/threonine protein phosphatase 1 and 2A inhibitors. Additionally, compounds of this type have displayed growth inhibition of various tumour cell lines. To further explore both of these inhibition pathways, a number of amide-acid norcantharidin analogues (15-26) were prepared. Compounds 23 and 24, containing two carboxylic acid residues, showed good PP1 and PP2A activity, with IC(50) values of approximately 15 and approximately 3 mum, respectively. Substituted aromatic amide analogues 45, 48, 49, 52, 53, and 54 also displayed good PP1 and PP2A inhibition, with IC(50) values in the range of 15-10 microM (PP1) and 11-5 microM (PP2A). However, bulky ortho substituents on the aromatic ring caused the aromatic ring to be skewed from the NCO planarity, leading to a decrease in PP1 and PP2A inhibition. A number of analogues, 20, 22, 25 and 46, showed excellent tumour growth inhibition, with 46 in particular being more potent than the lead, norcantharidin 2.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Cantaridina/análogos & derivados , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteína Fosfatasa 2/antagonistas & inhibidores , Animales , Antineoplásicos/química , Compuestos Bicíclicos Heterocíclicos con Puentes/síntesis química , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Cantaridina/síntesis química , Cantaridina/farmacología , Línea Celular , Cristalografía por Rayos X , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Humanos , Concentración 50 Inhibidora , Ratones , Proteína Fosfatasa 1/farmacología , Proteína Fosfatasa 2/farmacología , Relación Estructura-Actividad , Células Tumorales Cultivadas
18.
J Mol Biol ; 380(5): 789-98, 2008 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-18572192

RESUMEN

The apolipoprotein A5 gene (APOA5) is a key player in determining triglyceride concentrations in humans and mice. Since diabetes is often associated with hypertriglyceridemia, this study explores whether APOA5 gene expression is regulated by alteration in glucose homeostasis and the related pathways. d-Glucose activates APOA5 gene expression in a time- and dose-dependent manner in hepatocytes, and the glycolytic pathway involved was determined using d-glucose analogues and metabolites. Together, transient transfections, electrophoretic mobility shift assays and chromatin immunoprecipitation assays show that this regulation occurs at the transcriptional level through an increase of USF1/2 binding to an E-box in the APOA5 promoter. We show that this phenomenon is not due to an increase of mRNA or protein expression levels of USF. Using protein phosphatases 1 and 2A inhibitor, we demonstrate that d-glucose regulates the APOA5 gene via a dephosphorylation mechanism, resulting in an enhanced USF1/2-promoter binding. Last, subsequent suppressions of USF1/2 and phosphatases mRNA through siRNA gene silencing abolished the regulation. We demonstrate that the APOA5 gene is up regulated by d-glucose and USF through phosphatase activation. These findings may provide a new cross-talk between glucose and lipid metabolism.


Asunto(s)
Apolipoproteínas A/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Animales , Apolipoproteína A-V , Apolipoproteínas A/genética , Línea Celular , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Glucólisis , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Regiones Promotoras Genéticas , Unión Proteica , Proteína Fosfatasa 1/farmacología , Proteína Fosfatasa 2/farmacología , ARN Mensajero/metabolismo , Ratas , Factores de Tiempo , Transcripción Genética , Factores Estimuladores hacia 5'/metabolismo
19.
Cancer Res ; 68(12): 4658-65, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18559511

RESUMEN

Gain-of-function alterations to the sonic hedgehog (SHH) signaling cascade have been found in a wide range of tumors. Three SHH effectors, GLI1, GLI2, and GLI3, regulate transcription of diverse genes involved in cell growth and cell proliferation. Here, we show that protein phosphatase 2A (PP2A), its regulatory subunit, alpha4, and rapamycin, an inhibitor of the mammalian target of rapamycin kinase complex 1 (mTORC1), regulate the nuclear localization and transcriptional activity of GLI3. An increase in PP2A activity or treatment with rapamycin leads to cytosolic retention of GLI3 and, consequently, reduced transcription of the GLI3 target gene and cell cycle regulator, cyclin D1. Conversely, inhibition of PP2A results in increased expression of cyclin D1. In summary, our findings reveal the existence of a hitherto unrecognized molecular cross-talk between the oncogenic SHH pathway and the tumor suppressor PP2A and suggest a novel mechanism underlying the anticancerogenic effects of rapamycin.


Asunto(s)
Núcleo Celular/metabolismo , Inmunosupresores/farmacología , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteína Fosfatasa 2/farmacología , Sirolimus/farmacología , Proteínas Adaptadoras Transductoras de Señales , Ciclina D , Ciclinas/metabolismo , Citosol/metabolismo , Técnica del Anticuerpo Fluorescente , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Chaperonas Moleculares , Complejos Multiproteicos , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/metabolismo , Transporte de Proteínas , Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Fracciones Subcelulares , Serina-Treonina Quinasas TOR , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Transcripción Genética , Células Tumorales Cultivadas , Proteína con Dedos de Zinc GLI1 , Proteína Gli2 con Dedos de Zinc , Proteína Gli3 con Dedos de Zinc
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...