Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Psychol Med ; : 1-11, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38720515

RESUMEN

BACKGROUND: There is a clear demand for innovative therapeutics for bipolar disorder (BD). METHODS: We integrated the largest BD genome-wide association study (GWAS) dataset (NCase = 41 917, NControl = 371 549) with protein quantitative trait loci from brain, cerebrospinal fluid, and plasma. Using a range of integrative analyses, including Mendelian randomization (MR), Steiger filter analysis, Bayesian colocalization, and phenome-wide MR analysis, we prioritized novel drug targets for BD. Additionally, we incorporated data from the UK Biobank (NCase = 1064, NControl = 365 476) and the FinnGen study (NCase = 7006, NControl = 329 192) for robust biological validation. RESULTS: Through MR analysis, we found that in the brain, downregulation of DNM3, MCTP1, ABCB8 and elevation of DFNA5 and PDF were risk factors for BD. In cerebrospinal fluid, increased BD risk was associated with increased levels of FRZB, AGRP, and IL36A and decreased CTSF and LRP8. Plasma analysis revealed that decreased LMAN2L, CX3CL1, PI3, NCAM1, and TIMP4 correlated with increased BD risk, but ITIH1 did not. All these proteins passed Steiger filtering, and Bayesian colocalization confirmed that 12 proteins were colocalized with BD. Phenome-wide MR analysis revealed no significant side effects for potential drug targets, except for LRP8. External validation further underscored the concordance between the primary and validation cohorts, confirming MCTP1, DNM3, PDF, CTSF, AGRP, FRZB, LMAN2L, NCAM1, and TIMP4 are intriguing targets for BD. CONCLUSIONS: Our study identified druggable proteins for BD, including MCTP1, DNM3, and PDF in the brain; CTSF, AGRP, and FRZB in cerebrospinal fluid; and LMAN2L, NCAM1, and TIMP4 in plasma, delineating promising avenues to development of novel therapies.

2.
Chemphyschem ; 16(14): 2953-60, 2015 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-26266687

RESUMEN

Graphene shells with a controllable number of layers were directly synthesized on Cu nanoparticles (CuNPs) by chemical vapor deposition (CVD) to fabricate a graphene-encapsulated CuNPs (G/CuNPs) hybrid system for surface-enhanced Raman scattering (SERS). The enhanced Raman spectra of adenosine and rhodamine 6G (R6G) showed that the G/CuNPs hybrid system can strongly suppress background fluorescence and increase signal-to-noise ratio. In four different types of SERS systems, the G/CuNPs hybrid system exhibits more efficient SERS than a transferred graphene/CuNPs hybrid system and pure CuNPs and graphene substrates. The minimum detectable concentrations of adenosine and R6G by the G/CuNPs hybrid system can be as low as 10(-8) and 10(-10) M, respectively. The excellent linear relationship between Raman intensity and analyte concentration can be used for molecular detection. The graphene shell can also effectively prevent surface oxidation of Cu nanoparticles after exposure to ambient air and thus endow the hybrid system with a long lifetime. This work provides a basis for the fabrication of novel SERS substrates.


Asunto(s)
Cobre/química , Grafito/química , Nanopartículas del Metal , Espectrometría Raman/métodos , Microscopía de Fuerza Atómica , Propiedades de Superficie
3.
Front Immunol ; 15: 1444020, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39221247

RESUMEN

Electronic cigarettes (e-cigarettes) are thought to pose low risk of cancer because the components of e-cigarette liquid are not carcinogens. We analyzed the effects of the two major components, PG/VG and nicotine, on tumor development in preclinical models. We found that PG/VG promoted tumor cell migration in migration assays and contributed to more aggressive, metastatic, and immunosuppressive tumors in vivo, aggravated by the presence of nicotine. Whole body exposure of mice to PG/VG and nicotine rendered animals more susceptible to developing tumors with high frequencies of infiltrating proinflammatory macrophages expressing IL-6 and TNFα. Moreover, tumor-infiltrating and circulating T cells in e-cigarette exposed mice showed increased levels of immune checkpoints including CTLA4 and PD-1. Treatment with anti-CTLA4 antibody was able to abrogate metastasis with no detrimental effects on its ability to induce tumor regression in exposed mice. These findings suggest that the major components used in e-cigarette fluid can impact tumor development through induced immunosuppression.


Asunto(s)
Sistemas Electrónicos de Liberación de Nicotina , Nicotina , Animales , Ratones , Metástasis de la Neoplasia , Ratones Endogámicos C57BL , Línea Celular Tumoral , Antígeno CTLA-4/inmunología , Femenino , Movimiento Celular/efectos de los fármacos , Receptor de Muerte Celular Programada 1
4.
Cell Physiol Biochem ; 32(6): 1668-80, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24356372

RESUMEN

BACKGROUND/AIM: We have demonstrated that exogenous hydrogen sulfide (H2S) protects H9c2 cardiac cells against the doxorubicin (DOX)-induced injuries by inhibiting p38 mitogen-activated protein kinase (MAPK) pathway and that the p38 MAPK/nuclear factor-κB (NF-κB) pathway is involved in the DOX-induced inflammatory response and cytotoxicity. The present study attempts to test the hypothesis that exogenous H2S might protect cardiomyocytes against the DOX-induced inflammation and cytotoxicity through inhibiting p38 MAPK/NF-κB pathway. METHODS: H9c2 cardiac cells were exposed to 5µM DOX for 24 h to establish a model of DOX cardiotoxicity. The cells were pretreated with NaHS( a donor of H2S) or other drugs before exposure to DOX. Cell viability was analyzed by cell counter kit 8 ( CCK-8), The expression of NF-κB p65 and inducible nitric oxide synthase (iNOS) was detected by Western blot assay. The levels of interleukin-1ß (IL-1ß), IL-6 and tumor necrosis factor-α (TNF-α) were tested by enzyme-linked immunosorbent assay (ELISA). RESULTS: Our findings demonstrated that pretreatment of H9c2 cardiac cells with NaHS for 30 min before exposure to DOX markedly ameliorated the DOX-induced phosphorylation and nuclear translocation of NF-κB p65 subunit. Importantly, the pretreatment with NaHS significantly attenuated the p38 MAPK/NF-κB pathway-mediated inflammatory responses induced by DOX, as evidenced by decreases in the levels of IL-1ß, IL-6 and TNF-α. In addition, application of NaHS or IL-1ß receptor antagonist (IL-1Ra) or PDTC (an inhibitor of NF-κB) attenuated the DOX-induced expression of iNOS and production of nitric oxide (NO), respectively. Furthermore, IL-1Ra also dramatically reduced the DOX-induced cytotoxicity and phosphorylation of NF-κB p65. The pretreatment of H9c2 cells with N-acetyl-L-cysteine (NAC), a scavenger of reactive oxygen species (ROS) prior to exposure to DOX depressed the phosphorylation of NF-κB p65 induced by DOX. CONCLUSION: The present study has demonstrated the new mechanistic evidence that exogenous H2S attenuates the DOX-induced inflammation and cytotoxicity by inhibiting p38 MAPK/NF-κB pathway in H9c2 cardiac cells. We also provide novel data that the interaction between NF-κB pathway and IL-1ß is important in the induction of DOX-induced inflammation and cytotoxicity in H9c2 cardiac cells.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Doxorrubicina/toxicidad , Inflamación/inducido químicamente , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Sulfitos/farmacología , Acetilcisteína/farmacología , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Inflamación/patología , Interleucina-1beta/análisis , Interleucina-6/análisis , FN-kappa B/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación/efectos de los fármacos , Prolina/análogos & derivados , Prolina/farmacología , ARN Interferente Pequeño/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Tiocarbamatos/farmacología , Factor de Necrosis Tumoral alfa/análisis , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Anticancer Agents Med Chem ; 23(6): 717-725, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36221890

RESUMEN

BACKGROUND: Lung cancer is the leading cause of cancer-associated death and the first most diagnosed cancer in the world. More than 2 million new cases are diagnosed and 1.6 million people die due to lung cancer every year. It is urgent to explore novel drugs and approaches for lung cancer treatment. Cinobufotalin is a TCM isolated from dried toad venom, which has been used to treat lung cancer. However, the precise mechanism remains unclear. OBJECTIVE: This study was to investigate the mechanism of cinobufotalin treated in lung cancer. METHODS: Cell growth was identified by Cell Counting Kit-8 (CCK-8) assay. Besides, ferroptosis of lung cancer cells was determined by intracellular iron content, lactate dehydrogenase (LDH) release and mitochondrial membrane potential. Moreover, RNA levels and proteins were detected by quantitative reverse transcription-PCR (qRT-PCR) and Western blot (WB), respectively. In addition, the regulatory effect of hsa-miR-367-3p on TFRC was confirmed by luciferase reporter assay. RESULTS: This study indicated that cinobufotalin suppressed lung cancer cell growth through resibufogenin. Besides, cinobufotalin induced ferroptosis in lung cancer cells through resibufogenin. Moreover, cinobufotalin increased lncRNA LINC00597 level, whereas it downregulated hsa-miR-367-3p expression in lung cancer cells via resibufogenin. In addition, ferroptosis inducer transferrin receptor (TFRC) was the target of hsa-miR-367-3p, and lncRNA LINC00597 upregulates TFRC expression through sponging hsa-miR-367-3p in lung cancer cells. CONCLUSION: In summary, this study indicated that cinobufotalin induced ferroptosis to suppress lung cancer cell growth by lncRNA LINC00597/hsa-miR-367-3p/TFRC pathway via resibufogenin might provide novel therapeutic targets for lung cancer therapy.


Asunto(s)
Ferroptosis , Neoplasias Pulmonares , MicroARNs , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , MicroARNs/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Receptores de Transferrina , Proliferación Celular
6.
Mult Scler Relat Disord ; 70: 104484, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36608538

RESUMEN

BACKGROUND: Adequate response to the SARS-CoV-2 vaccine represents an important treatment goal in caring for patients with multiple sclerosis (MS) during the ongoing COVID-19 pandemic. Previous data so far have demonstrated lower spike-specific IgG responses following two SARS-CoV-2 vaccinations in MS patients treated with sphingosine-1-phosphate (S1P) receptor modulators and anti-CD20 monoclonal antibodies (mAb) compared to other disease modifying therapies (DMTs). It is unknown whether subsequent vaccinations can augment antibody responses in these patients. OBJECTIVES: The goal of this observational study was to determine the effects of a third SARS-CoV-2 vaccination on antibody and T cell responses in MS patients treated with anti-CD20 mAb or S1P receptor modulators. METHODS: Vaccine responses in patients treated with anti-CD20 antibodies (ocrelizumab and ofatumumab) or S1P receptor modulators (fingolimod and siponimod) were evaluated before and after third SARS-CoV-2 vaccination as part of an ongoing longitudinal study. Total spike protein and spike receptor binding domain (RBD)-specific IgG responses were measured by Luminex bead-based assay. Spike-specific CD4+ and CD8+ T cell responses were measured by activation-induced marker expression. RESULTS: MS patients and healthy controls were enrolled before and following SARS-CoV-2 vaccination. A total of 31 MS patients (n = 10 ofatumumab, n = 13 ocrelizumab, n = 8 S1P) and 10 healthy controls were evaluated through three SARS-CoV-2 vaccinations. Compared to healthy controls, total spike IgG was significantly lower in anti-CD20 mAb-treated patients and spike RBD IgG was significantly lower in anti-CD20 mAb and S1P-treated patients following a third vaccination. While seropositivity was 100% in healthy controls after a third vaccination, total spike IgG and spike RBD IgG seropositivity were lower in ofatumumab (60% and 60%, respectively), ocrelizumab (85% and 46%, respectively), and S1P-treated patients (100% and 75%, respectively). Longer treatment duration, including prior treatment history, appeared to negatively impact antibody responses. Spike-specific CD4+ and CD8+ T cell responses were well maintained across all groups following a third vaccination. Finally, immune responses were also compared in patients who were vaccinated prior to or following ofatumumab treatment. Antibody responses were significantly higher in those patients who received their primary SARS-CoV-2 vaccination prior to initiating ofatumumab treatment. CONCLUSIONS: This study adds to the evolving understanding of SARS-CoV-2 vaccine responses in people with MS treated with disease-modifying therapies (DMTs) known to suppress humoral immunity. Our findings provide important information for optimizing vaccine immunity in at-risk MS patient populations.


Asunto(s)
COVID-19 , Esclerosis Múltiple , Moduladores de los Receptores de fosfatos y esfingosina 1 , Humanos , Inmunidad Humoral , Vacunas contra la COVID-19 , Receptores de Esfingosina-1-Fosfato , SARS-CoV-2 , Estudios Longitudinales , Pandemias , Vacunación , Anticuerpos Monoclonales , Inmunoglobulina G , Anticuerpos Antivirales
7.
Mol Cell Biochem ; 363(1-2): 419-26, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22203419

RESUMEN

The roles of hydrogen sulfide (H(2)S) and endoplasmic reticulum (ER) stress in doxorubicin (DOX)-induced cardiotoxicity are still unclear. This study aimed to dissect the hypothesis that H(2)S could protect H9c2 cells against DOX-induced cardiotoxicity by inhibiting ER stress. Our results showed that exposure of H9c2 cells to DOX significantly inhibited the expression and activity of cystathionine-γ-lyase (CSE), a synthetase of H(2)S, accompanied by the decreased cell viability and the increased reactive oxygen species (ROS) accumulation. In addition, exposure of cells to H(2)O(2) (an exogenous ROS) mimicked the inhibitory effect of DOX on the expression and activity of CSE. Pretreatment with N-acetyl-L: -cysteine (NAC) (a ROS scavenger) attenuated intracellular ROS accumulation, cytotoxicity, and the inhibition of expression and activity of CSE induced by DOX. Notably, the ER stress-related proteins, including glucose-regulated protein 78 (GRP78) and C/EBP homologous protein (CHOP) were obviously upregulated in DOX-treated H9c2 cells. Pretreatment with sodium hydrosulfide (NaHS, a H(2)S donor) before DOX exposure markedly suppressed DOX-induced overexpressions of GRP78 and CHOP, cytotoxicity and oxidative stress. In conclusion, we have demonstrated that ROS-mediated inhibition of CSE is involved in DOX-induced cytotoxicity in H9c2 cells, and that exogenous H(2)S can confer protection against DOX-induced cardiotoxicity partly through inhibition of ER stress.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Antioxidantes/farmacología , Doxorrubicina/toxicidad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Sulfuro de Hidrógeno/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Sulfuros/farmacología , Acetilcisteína/farmacología , Animales , Antioxidantes/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cistationina gamma-Liasa/metabolismo , Citoprotección , Relación Dosis-Respuesta a Droga , Depuradores de Radicales Libres/farmacología , Proteínas de Choque Térmico/metabolismo , Peróxido de Hidrógeno/toxicidad , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Oxidantes/toxicidad , Ratas , Especies Reactivas de Oxígeno/metabolismo , Sulfuros/metabolismo , Factores de Tiempo , Factor de Transcripción CHOP/metabolismo
8.
Mol Cell Biochem ; 362(1-2): 149-57, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22134701

RESUMEN

Hydrogen sulfide (H(2)S) has been shown to exert cardioprotective effects. However, the roles of extracellular signal-regulated protein kinases 1/2 (ERK1/2) in H(2)S-induced cardioprotection have not been completely elucidated. In this study, cobalt chloride (CoCl(2)), a chemical hypoxia mimetic agent, was applied to treat H9c2 cells to establish a chemical hypoxia-induced cardiomyocyte injury model. The results showed that pretreatment with NaHS (a donor of H(2)S) before exposure to CoCl(2) attenuated the decreased cell viability, the increased apoptosis rate, the loss of mitochondrial membrane potential (ΔΨm), and the intracellular accumulation of reactive oxygen species (ROS) in H9c2 cells. Exposure of H9c2 cells to CoCl(2) or hydrogen peroxide (H(2)O(2)) upregulated expression of phosphorylated (p) ERK1/2, which was reduced by pretreatment with NaHS or N-acetyl-L-cysteine, a ROS scavenger. More importantly, U0126, a selective inhibitor of ERK1/2, mimicked the above cytoprotection of H(2)S against CoCl(2)-induced injury in H9c2 cells. In conclusion, these results indicate that H(2)S protects H9c2 cells against chemical hypoxia-induced injury partially by inhibiting ROS-mediated activation of ERK1/2.


Asunto(s)
Hipoxia de la Célula , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sulfuro de Hidrógeno/farmacología , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Animales , Apoptosis/efectos de los fármacos , Butadienos/farmacología , Cardiotónicos/farmacología , Hipoxia de la Célula/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cobalto/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Peróxido de Hidrógeno/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Nitrilos/farmacología , Ratas
9.
Clin Exp Pharmacol Physiol ; 38(1): 42-9, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21083699

RESUMEN

1. Increasing evidence indicates that hydrogen sulphide (H2S) may serve as an important biological cytoprotective agent. Heat shock protein (Hsp) 90 can attenuate stress-induced injury. However, whether Hsp90 mediates the cytoprotective effect of H2S against chemical hypoxia-induced injury in PC12 cells is not known. 2. In the present study, CoCl2 (a chemical hypoxia mimetic) was used to treat PC12 cells to create a model of chemical hypoxia. To explore the role of Hsp90 in the cytoprotection afforded by H2S against chemical hypoxia-induced injury, 2 µmol/L 17-allylaminogeldanamycin (17-AAG), a selective inhibitor of Hsp90, was administered for 30 min prior to preconditioning with 400 µmol/L NaHS, followed by chemical hypoxia. 3. Cobalt chloride reduced cell viability (by 52.7 ± 1.5%), increased PC12 cell apoptosis (by 42.1 ± 1.5%), induced reactive oxygen species (ROS) by 3.79% compared with control and induced the dissipation of mitochondrial membrane potential (MMP) by 2.56% compared with control. 4. Pretreatment of PC12 cells with 100-400 µmol/L sodium hydrosulphide (NaHS), an H2S donor, for 3 h prior to exposure to 600 µmol/L CoCl2 provided significant, concentration-dependant protection to PC12 cells against CoCl2-induced cytotoxicity. Specifically, pretreatment of PC12 cells with 400 µmol/L NaHS decreased apoptosis to 16.77 ± 1.77% and blocked the CoCl2-induced increase in ROS production and loss of MMP. 5. At 400 µmol/L, NaHS upregulated Hsp90 in a time-dependant manner (over the period 0-180 min). In addition to its effects on Hsp90 expression, NaHS pretreatment of PC12 cells augmented the overexpression of Hsp90 induced by 600 µmol/L CoCl2 by 1.38-fold (P < 0.01). 6. Treatment of PC12 cells with 2 µmol/L 17-AAG for 30 min prior to NaHS pretreatment blocked the overexpression of Hsp90 induced by NaHS preconditioning, as evidenced by decreased cell viability (by 54.2 + 1.2%; P < 0.01), increased PC12 cell apoptosis (by 36.6 ± 1.2%; P < 0.01) and increasing ROS production. 7. The findings of the present study provide novel evidence that Hsp90 mediates H2S-induced neuroprotection against chemical hypoxia-induced injury via anti-oxidant and anti-apoptotic effects.


Asunto(s)
Apoptosis/efectos de los fármacos , Citoprotección/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/fisiología , Sulfuro de Hidrógeno/farmacología , Hipoxia/complicaciones , Animales , Antioxidantes/farmacología , Hipoxia de la Célula/efectos de los fármacos , Cobalto , Citotoxinas , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Proteínas HSP90 de Choque Térmico/metabolismo , Hipoxia/inducido químicamente , Hipoxia/metabolismo , Hipoxia/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Células PC12 , Ratas , Especies Reactivas de Oxígeno/metabolismo
10.
Brain Inj ; 25(6): 604-13, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21534738

RESUMEN

PRIMARY OBJECTIVE: Recent evidence suggests that delayed hypoxic post-conditioning is neuroprotective. The aim of the present study was to test whether early post-conditioning applied immediately after hypoxia could protect cultured neurons from hypoxia/reoxygenation (H/R)-induced injuries. METHODS: Primary cortical neuronal culture depleted of microglia was exposed to H/R. Post-conditioning started immediately after hypoxia and consisted of three cycles of 15-minutes of reoxygenation and 15-minutes of hypoxia. Cell viability assay was performed using Cell Counting Kit-8 (CCK-8). Apoptosis was evaluated by Hoechst 33258 staining, FITC-Annexin V/PI double staining and Western blot assay (testing the cleaved caspase-3 expression). Reactive oxygen species (ROS), intracellular Ca(2+) and mitochondrial membrane potential (MMP) were examined using confocal laser-scanning microscopy. MAIN RESULTS: H/R significantly reduced cell viability and increased neuronal apoptosis and necrosis. Furthermore, the expression of cleaved caspase-3, ROS production and intracellular Ca(2+) were increased. MMP was attenuated. Injuries induced by H/R were substantially attenuated by early hypoxic post-conditioning. Changes in cleaved caspase-3 expression, ROS production, intracellular Ca(2+) level and MMP in response to H/R were significantly decreased by the post-conditioning. CONCLUSIONS: The findings demonstrated that early hypoxic post-conditioning could protect neurons against H/R-induced injuries independent of microglial cells, possibly by inhibiting ROS over-production and intracellular Ca(2+) accumulation and maintaining MMP.


Asunto(s)
Apoptosis/fisiología , Hipoxia de la Célula/fisiología , Supervivencia Celular/fisiología , Corteza Cerebral/metabolismo , Neuronas/fisiología , Especies Reactivas de Oxígeno/metabolismo , Animales , Western Blotting , Células Cultivadas , Corteza Cerebral/fisiopatología , Ratas , Ratas Sprague-Dawley
11.
Clin Exp Pharmacol Physiol ; 37(3): 316-21, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19769612

RESUMEN

1. The aim of the present study was to investigate the effect of hydrogen sulphide (H(2)S) on cobalt chloride (CoCl(2))-induced injury in H9c2 embryonic rat cardiac cells. 2. After 36 h incubation in the presence of 600 micromol/L CoCl(2), reduced cell viability of H9c2 cells was observed, as well as the induction of apoptosis. In addition, CoCl(2) (600 micromol/L) enhanced the production of reactive oxygen species (ROS) and the expression of cleaved caspase 3, induced a loss of mitochondrial membrane potential (MMP) and decreased reduced glutathione (GSH) production. These results suggest that CoCl(2) induces similar responses to hypoxia/ischaemia. 3. Pretreatment of cells with 400 micromol/L NaHS (a H(2)S donor) for 30 min prior to exposure to CoCl(2) (600 micromol/L) significantly protected H9c2 cells against CoCl(2)-induced injury. Specifically, increased cell viability and decreased apoptosis were observed. In addition, NaHS pretreatment blocked the CoCl(2)-induced increases in ROS production and cleaved caspase 3 expression, as well as the decreases in GSH production and loss of MMP. 4. Pretreatment of cells with 2000 micromol/L N-acetylcysteine (NAC), a ROS scavenger, for 1 h prior to CoCl(2) exposure significantly protected H9c2 cells against CoCl(2)-induced injury, specifically enhancing cell viability, decreasing ROS production and preventing loss of MMP. 5. The findings of the present study suggest that H(2)S protects H9c2 cells against CoCl(2)-induced injury by suppressing oxidative stress and caspase 3 activation.


Asunto(s)
Cobalto/toxicidad , Citoprotección/efectos de los fármacos , Citoprotección/fisiología , Sulfuro de Hidrógeno/farmacología , Animales , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Miocardio/citología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Ratas , Especies Reactivas de Oxígeno/metabolismo
12.
Nat Med ; 26(1): 118-130, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31873312

RESUMEN

Gene silencing with virally delivered shRNA represents a promising approach for treatment of inherited neurodegenerative disorders. In the present study we develop a subpial technique, which we show in adult animals successfully delivers adeno-associated virus (AAV) throughout the cervical, thoracic and lumbar spinal cord, as well as brain motor centers. One-time injection at cervical and lumbar levels just before disease onset in mice expressing a familial amyotrophic lateral sclerosis (ALS)-causing mutant SOD1 produces long-term suppression of motoneuron disease, including near-complete preservation of spinal α-motoneurons and muscle innervation. Treatment after disease onset potently blocks progression of disease and further α-motoneuron degeneration. A single subpial AAV9 injection in adult pigs or non-human primates using a newly designed device produces homogeneous delivery throughout the cervical spinal cord white and gray matter and brain motor centers. Thus, spinal subpial delivery in adult animals is highly effective for AAV-mediated gene delivery throughout the spinal cord and supraspinal motor centers.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Dependovirus/metabolismo , Silenciador del Gen , Técnicas de Transferencia de Gen , Neuronas Motoras/patología , Degeneración Nerviosa/terapia , Piamadre/patología , Médula Espinal/patología , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Atrofia , Progresión de la Enfermedad , Potenciales Evocados Motores , Femenino , Regulación de la Expresión Génica , Humanos , Inflamación/patología , Interneuronas/patología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Desarrollo de Músculos , Degeneración Nerviosa/genética , Degeneración Nerviosa/fisiopatología , Piamadre/fisiopatología , Primates , Pliegue de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/administración & dosificación , Médula Espinal/diagnóstico por imagen , Médula Espinal/fisiopatología , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Porcinos
13.
Clin Exp Pharmacol Physiol ; 36(3): 304-11, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18986327

RESUMEN

1. Cytoprotection by H(2)O(2) preconditioning against oxidative stress-induced apoptosis of PC12 cells has been demonstrated previously. In the present study, we investigated the effects of H(2)O(2) preconditioning on nuclear factor (NF)-kappaB activation and the role of NF-kappaB in the adaptive cytoprotection of H(2)O(2) preconditioning in PC12 cells. 2. The PC12 cells were preconditioned with 100 micromol/L H(2)O(2) for 90 min, followed by 24 h recovery and subsequent exposure to 300 micromol/L H(2)O(2) for a further 12 h. 3. The results showed that preconditioning with 100 micromol/L H(2)O(2) upregulated NF-kappaB expression and enhanced its nuclear translocation and DNA binding activity. In addition to its own effects on NF-kappaB expression, H(2)O(2) preconditioning also promoted the overexpression of NF-kappaB induced by a lethal concentration of H(2)O(2) (300 micromol/L). 4. N-Tosyl-l-phenylalanine chloromethyl ketone (TPCK; 20 micromol/L), an inhibitor of NF-kappaB, was administered 20 min before preconditioning with 100 micromol/L H(2)O(2). At this concenteration, TPCK blocked the overexpression of NF-kappaB induced by H(2)O(2) preconditioning, accompanied by attenuation of H(2)O(2) preconditioning-induced cytoprotection. The inhibition of NF-kappaB by TPCK enhanced caspase 3 activity induced by 300 micromol/L H(2)O(2). 5. The findings of the present study provide novel evidence for the effects of preconditioning with H(2)O(2) on constitutive activation of NF-kappaB, which contributes to the adaptive cytoprotection of H(2)O(2) preconditioning against PC12 cells apoptosis.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/metabolismo , Apoptosis/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Oxidantes/farmacología , Estrés Oxidativo/efectos de los fármacos , Feocromocitoma/metabolismo , Factor de Transcripción ReIA/metabolismo , Transporte Activo de Núcleo Celular , Neoplasias de las Glándulas Suprarrenales/patología , Animales , Citoprotección , ADN/metabolismo , Relación Dosis-Respuesta a Droga , Peróxido de Hidrógeno/toxicidad , Oxidantes/toxicidad , Células PC12 , Feocromocitoma/patología , Ratas , Factores de Tiempo , Clorometilcetona de Tosilfenilalanila/farmacología , Factor de Transcripción ReIA/antagonistas & inhibidores
14.
Brain Behav Immun ; 22(1): 114-23, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17919885

RESUMEN

We have previously demonstrated that activation of p38 mitogen-activated protein kinase (p38 MAPK) in the spinal microglia mediates morphine antinociceptive tolerance. Minocycline, a selective inhibitor of microglia activation, has been reported to attenuate peripheral inflammation-induced hyperalgesia by depressing p38 MAPK in the spinal microglia. The aim of the present study is to explore the effect of intrathecal minocycline on the development of morphine antinociceptive tolerance and p38 activation in the spinal microglia induced by chronic morphine treatment. Minocycline (20, 50 and 100 microg) was given intrathecally 30 min before each morphine (15 microg) administration for consecutive 7 days. It was shown that minocycline attenuated tolerance to morphine analgesia in a dose-dependent manner. Minocycline administration (50 microg) which was initiated on day 4 followed by another 4 days administration partially reversed the established morphine antinociceptive tolerance. However, minocycline treatment which was started on day 8 followed by its administration for 4 more days failed to reverse the established morphine tolerance. Immunohistochemical analysis showed that chronic intrathecal morphine-induced activation of p38 MAPK in the spinal microglia. Minocycline at a dose that was shown to antagonize tolerance to morphine analgesia significantly inhibited the increase in p38 MAPK activation in the spinal microglia. To our knowledge, this is the first study to demonstrate that minocycline antagonizes morphine antinociceptive tolerance, possibly due to the inhibition of p38 activation in the spinal microglia.


Asunto(s)
Analgésicos Opioides/farmacología , Microglía/enzimología , Minociclina/farmacología , Morfina/farmacología , Nociceptores/efectos de los fármacos , Médula Espinal/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/antagonistas & inhibidores , Animales , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Tolerancia a Medicamentos , Activación Enzimática/efectos de los fármacos , Inmunohistoquímica , Inyecciones Espinales , Masculino , Minociclina/administración & dosificación , Morfina/administración & dosificación , Morfina/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Médula Espinal/citología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Sci Transl Med ; 10(440)2018 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-29743351

RESUMEN

The use of autologous (or syngeneic) cells derived from induced pluripotent stem cells (iPSCs) holds great promise for future clinical use in a wide range of diseases and injuries. It is expected that cell replacement therapies using autologous cells would forego the need for immunosuppression, otherwise required in allogeneic transplantations. However, recent studies have shown the unexpected immune rejection of undifferentiated autologous mouse iPSCs after transplantation. Whether similar immunogenic properties are maintained in iPSC-derived lineage-committed cells (such as neural precursors) is relatively unknown. We demonstrate that syngeneic porcine iPSC-derived neural precursor cell (NPC) transplantation to the spinal cord in the absence of immunosuppression is associated with long-term survival and neuronal and glial differentiation. No tumor formation was noted. Similar cell engraftment and differentiation were shown in spinally injured transiently immunosuppressed swine leukocyte antigen (SLA)-mismatched allogeneic pigs. These data demonstrate that iPSC-NPCs can be grafted into syngeneic recipients in the absence of immunosuppression and that temporary immunosuppression is sufficient to induce long-term immune tolerance after NPC engraftment into spinally injured allogeneic recipients. Collectively, our results show that iPSC-NPCs represent an alternative source of transplantable NPCs for the treatment of a variety of disorders affecting the spinal cord, including trauma, ischemia, or amyotrophic lateral sclerosis.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/trasplante , Médula Espinal/trasplante , Envejecimiento , Animales , Diferenciación Celular , Reprogramación Celular , Enfermedad Crónica , Fibroblastos/citología , Regulación de la Expresión Génica , Tolerancia Inmunológica , Inmunidad Humoral , Terapia de Inmunosupresión , Neostriado/patología , Células-Madre Neurales/citología , Neuronas/citología , Ratas , Piel/citología , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/terapia , Análisis de Supervivencia , Porcinos , Porcinos Enanos , Trasplante Homólogo , Trasplante Isogénico
17.
Brain Res ; 1069(1): 235-43, 2006 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-16403466

RESUMEN

Compelling evidence has suggested that spinal glial cells were activated by chronic morphine treatment and involved in the development of morphine tolerance. However, the mechanisms of glial activation were still largely unknown in morphine tolerance. In present study, we investigated the role of p38 mitogen-activated protein kinase (p38 MAPK) in the spinal cord in the development of chronic morphine antinociceptive tolerance. We found that intrathecal administration of morphine (15 microg) daily for 7 consecutive days significantly induced an increase in number of phospho-p38 (p-p38) immunoreactive cells in the spinal cord compared with chronic saline or acute morphine treated rats. Double immunofluorescence staining revealed that p-p38 immunoreactivity was exclusively restricted in the activated spinal microglia, not in astrocytes or neurons. Repeated intrathecal administration of 4-(4-fluorophenyl)-2-(4-methylsulfonylphenyl)-5-(4-pyridyl)-1H-imidazole (SB203580) (10 microg or 2 microg), a specific p38 inhibitor, 30 min before each morphine injection for 7 consecutive days significantly attenuated tolerance to morphine analgesia assessed by tail flick test. However, a single intrathecal administration of SB203580 (10 microg) did not antagonize the established tolerance to morphine analgesia. Taken together, these findings suggested that p38 MAPK activation in the spinal microglia was involved in the development of morphine antinociceptive tolerance. Inhibition of p38 MAPK by SB203580 in the spinal cord attenuated but not reversed the tolerance to morphine analgesia. The present study provides the first evidence that p38 activation in spinal microglia played an important role in the development of tolerance to morphine analgesia.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Microglía/fisiología , Dependencia de Morfina , Morfina/administración & dosificación , Médula Espinal/patología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Analgésicos Opioides/efectos adversos , Análisis de Varianza , Animales , Conducta Animal , Antígeno CD11b/metabolismo , Recuento de Células/métodos , Esquema de Medicación , Interacciones Farmacológicas , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Proteína Ácida Fibrilar de la Glía/metabolismo , Imidazoles/farmacología , Inmunohistoquímica/métodos , Masculino , Morfina/efectos adversos , Dependencia de Morfina/metabolismo , Dependencia de Morfina/patología , Dependencia de Morfina/fisiopatología , Fosfopiruvato Hidratasa/metabolismo , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
18.
Neurosci Lett ; 410(3): 174-7, 2006 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-17101217

RESUMEN

We have demonstrated that the activation of p38 mitogen-activated protein kinase (MAPK) in the spinal microglia played an essential role in the development of morphine antinociceptive tolerance. The aim of this study was to investigate whether inhibition of neuronal nitric oxide synthase (nNOS) attenuated tolerance to morphine analgesia by modulating p38 activation in the spinal microglia. It was shown that the selective inhibitor of nNOS, 7-NINA (7-Nitroindazole, sodium salt) (25 microg, i.t.) attenuated not only the development of morphine antinociceptive tolerance, but also the activation of p38 MAPK in the spinal microglia induced by chronic intrathecal administration of morphine. Our results suggest that neuronal NO signals to microglia, leading to the upregulation of microglial phospho-p38 MAPK. Such p38 MAPK activation in microglia is consistent with a potential role in the development of morphine antinociceptive tolerance. We demonstrated for the first time that the inhibition of nNOS attenuated morphine antinociceptive tolerance by reducing p38 MAPK activation in the spinal microglia.


Asunto(s)
Microglía/efectos de los fármacos , Morfina/administración & dosificación , Narcóticos/administración & dosificación , Óxido Nítrico Sintasa de Tipo I/fisiología , Médula Espinal/citología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Análisis de Varianza , Animales , Recuento de Células/métodos , Interacciones Farmacológicas , Tolerancia a Medicamentos/fisiología , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Técnica del Anticuerpo Fluorescente/métodos , Indazoles/farmacología , Inyecciones Espinales/métodos , Masculino , Ratas , Ratas Sprague-Dawley
19.
Life Sci ; 79(9): 870-6, 2006 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-16616209

RESUMEN

The induction of inducible nitric oxide synthase (iNOS) in response to different stress is associated with simultaneous induction of cyclooxygenase-2 (COX-2) in various cell types. Both iNOS and COX-2 have been reported to mediate the late phase of cardioprotection induced by different preconditioning. However, whether both iNOS and COX-2 are mediators in the neuroprotection induced by preconditioning with hydrogen peroxide (H(2)O(2)) at low concentration is unknown. In this study, using the neurosecretory cell line-PC12 cells to set up the model of neuroprotection of preconditioning with H(2)O(2) against apoptosis, we first investigate what changes in expression of iNOS and COX-2 appear during H(2)O(2) preconditioning, then determine if both iNOS inhibitor and COX-2 inhibitor interfere with the neuroprotection elicited by preconditioning with H(2)O(2). We found that preconditioning with H(2)O(2) at 10 microM significantly protected PC12 cells against apoptosis induced by lethal H(2)O(2) (50 microM) and increased the expression of iNOS and COX-2 and that selective iNOS inhibitor, aminoguanidine (AG) and COX-2 inhibitor, NS-398 obviously blocked the protective effects induced by preconditioning with 10 microM H(2)O(2). The results of this study suggest that both iNOS and COX-2 are mediators of the neuroprotection induced by preconditioning with oxidative stress (H(2)O(2) at low concentration) in PC12 cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclooxigenasa 2/biosíntesis , Peróxido de Hidrógeno/farmacología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Estrés Oxidativo/fisiología , Animales , Western Blotting , Núcleo Celular/ultraestructura , Supervivencia Celular/fisiología , Colorantes , Inhibidores de la Ciclooxigenasa 2/farmacología , Citometría de Flujo , Nitrobencenos/farmacología , Células PC12 , Ratas , Sulfonamidas/farmacología
20.
Brain Res ; 1057(1-2): 57-64, 2005 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-16129420

RESUMEN

The present study is designed to investigate the effects of preconditioning with different doses of hydrogen peroxide (H2O2) on oxidative stress-induced apoptosis and the changes in mitochondrial membrane potential (MMP), intracellular reactive oxygen species (ROS) level, and expression of Bcl-2 during H2O2 preconditioning in rat pheochromocytoma (PC12) cells. It was shown that (1) H2O2 induced apoptosis in PC12 cells in a dose-dependent manner; (2) the preconditioning with 10 micromol L(-1) or 20 micromol L(-1) H2O2 can significantly protect PC12 cells against apoptosis induced by 50 or 100 micromol L(-1) H2O2, low (5 micromol L(-1)) and higher (30 micromol L(-1)) concentrations of H2O2 had no cytoprotections; (3) high concentration (100 micromol L(-1)) of H2O2 reduced MMP and expression of Bcl-2, and increased ROS level, but these effects were blocked by preconditioning with 10 micromol L(-1) H2O2; (4) the preconditioning with 10 micromol L(-1) H2O2 induced overexpression of Bcl-2. These results suggested that the preconditioning with low dose of H2O2 could protect the oxidative stress-induced PC12 cells apoptosis not only by preventing the reduction of MMP and expression of Bcl-2 as well as increase in ROS level, but also through overexpression of Bcl-2. It was indicated that overexpression of Bcl-2 may play a key role in the cytoprotection induced by preconditioning with low dose of H2O2 in PC12 cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Potenciales de la Membrana/efectos de los fármacos , Estrés Oxidativo/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Apoptosis/fisiología , Western Blotting/métodos , Recuento de Células/métodos , Muerte Celular , Relación Dosis-Respuesta a Droga , Citometría de Flujo/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Inmunohistoquímica/métodos , Potenciales de la Membrana/fisiología , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/fisiología , Estrés Oxidativo/efectos de los fármacos , Células PC12 , Ratas , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA