Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 298(4): 101817, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35278434

RESUMEN

Expression of the receptor tyrosine kinase ephrin receptor A10 (EphA10), which is undetectable in most normal tissues except for the male testis, has been shown to correlate with tumor progression and poor prognosis in several malignancies, including triple-negative breast cancer (TNBC). Therefore, EphA10 could be a potential therapeutic target, likely with minimal adverse effects. However, no effective clinical drugs against EphA10 are currently available. Here, we report high expression levels of EphA10 in tumor regions of breast, lung, and ovarian cancers as well as in immunosuppressive myeloid cells in the tumor microenvironment. Furthermore, we developed anti-EphA10 monoclonal antibodies (mAbs) that specifically recognize cell surface EphA10, but not other EphA family isoforms, and target tumor regions precisely in vivo with no apparent accumulation in other organs. In syngeneic TNBC mouse models, we found that anti-EphA10 mAb clone #4 enhanced tumor regression, therapeutic response rate, and T cell-mediated antitumor immunity. Notably, the chimeric antigen receptor T cells derived from clone #4 significantly inhibited TNBC cell viability in vitro and tumor growth in vivo. Together, our findings suggest that targeting EphA10 via EphA10 mAbs and EphA10-specific chimeric antigen receptor-T cell therapy may represent a promising strategy for patients with EphA10-positive tumors.


Asunto(s)
Anticuerpos Monoclonales , Receptores Quiméricos de Antígenos , Receptores de la Familia Eph , Linfocitos T , Neoplasias de la Mama Triple Negativas , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Ratones , Receptores de la Familia Eph/inmunología , Linfocitos T/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Int J Mol Sci ; 23(19)2022 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-36233084

RESUMEN

In addition to the fundamental role of insulin-like growth factor (IGF)/IGF-1 receptor (IGF-1R) signaling dysregulation in cancer initiation and proliferation, the IGF/IGF-1R signaling also plays an important role in the maintenance of stem cell characteristics and enhancement of stem cell-based therapeutic efficacy. This review focused on the role of IGF/IGF-1R signaling in preclinical IGF-targeted therapies, including IGF-1R monoclonal antibodies, IGF-1R tyrosine kinase inhibitors, and neutralizing antibodies of IGFs in multiple tumors and endocrine disorders. On the other hand, the function of IGF/IGF-1R signaling in stem cell self-renewal, pluripotency and therapeutic efficacy in regenerative medicine was outlined. Finally, the review summarized ongoing studies on IGF/IGF-1R signaling blockade in multiple cancers and highlighted the IGF-1R signaling modifications in stem cells as a potential strategy to improve stem cell-based therapeutics in regenerative medicine.


Asunto(s)
Neoplasias , Somatomedinas , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Línea Celular Tumoral , Humanos , Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptor IGF Tipo 1/metabolismo , Medicina Regenerativa , Somatomedinas/uso terapéutico , Células Madre/metabolismo
3.
J Gastroenterol Hepatol ; 36(7): 1988-1996, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33462840

RESUMEN

BACKGROUND AND AIM: Hepatocellular carcinoma (HCC) remains a serious cause of cancer-related deaths worldwide. Developing new therapeutic strategies is urgently needed to improve the outcomes of HCC patients. Dendritic cell (DC)-based vaccines and programmed death 1 (PD-1) immune checkpoint inhibitors have been regarded as potential immunotherapeutics for HCC. However, the therapeutic efficacy of combining these two treatments for HCC remains to be evaluated. METHODS: In this study, DCs were derived from mouse bone marrow and pulsed with mouse HCC cell lysates to generate a DC vaccine. A monoclonal antibody that blocks the interaction of mouse PD-1 with its ligands was used as a PD-1 inhibitor. An orthotopic HCC mouse model was established to assess the effect of a DC vaccine in combination with a PD-1 inhibitor on overall survival and tumor volume. RESULTS: Compared with the untreated control, single treatment with a DC vaccine or PD-1 inhibitor prolonged the overall survival and reduced the tumor volume of HCC mice. Further, compared with the single treatment with the DC vaccine or the PD-1 inhibitor, a combination treatment using both agents elicited a higher cytotoxicity of T cells against HCC cells and resulted in a better overall survival, smaller tumor volume, and greater tumor cell apoptosis in HCC mice. CONCLUSIONS: Our results suggest that a combination treatment with DC vaccine and PD-1 inhibitor may be a promising therapeutic strategy for HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Vacunas , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Células Dendríticas/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico , Neoplasias Hepáticas/tratamiento farmacológico , Ratones , Receptor de Muerte Celular Programada 1 , Vacunas/uso terapéutico
4.
Int J Mol Sci ; 22(19)2021 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-34638579

RESUMEN

Parkinson's disease (PD) is a degenerative disease that can cause motor, cognitive, and behavioral disorders. The treatment strategies being developed are based on the typical pathologic features of PD, including the death of dopaminergic (DA) neurons in the substantia nigra of the midbrain and the accumulation of α-synuclein in neurons. Peiminine (PMN) is an extract of Fritillaria thunbergii Miq that has antioxidant and anti-neuroinflammatory effects. We used Caenorhabditis elegans and SH-SY5Y cell models of PD to evaluate the neuroprotective potential of PMN and address its corresponding mechanism of action. We found that pretreatment with PMN reduced reactive oxygen species production and DA neuron degeneration caused by exposure to 6-hydroxydopamine (6-OHDA), and therefore significantly improved the DA-mediated food-sensing behavior of 6-OHDA-exposed worms and prolonged their lifespan. PMN also diminished the accumulation of α-synuclein in transgenic worms and transfected cells. In our study of the mechanism of action, we found that PMN lessened ARTS-mediated degradation of X-linked inhibitor of apoptosis (XIAP) by enhancing the expression of PINK1/parkin. This led to reduced 6-OHDA-induced apoptosis, enhanced activity of the ubiquitin-proteasome system, and increased autophagy, which diminished the accumulation of α-synuclein. The use of small interfering RNA to down-regulate parkin reversed the benefits of PMN in the PD models. Our findings suggest PMN as a candidate compound worthy of further evaluation for the treatment of PD.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Cevanas/farmacología , Enfermedad de Parkinson/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , alfa-Sinucleína/metabolismo , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans/metabolismo , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Degeneración Nerviosa/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Sustancia Negra/metabolismo , Ubiquitina/metabolismo
5.
Int J Mol Sci ; 21(12)2020 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-32585871

RESUMEN

The movement disorder Parkinson's disease (PD) is the second most frequently diagnosed neurodegenerative disease, and is associated with aging, the environment, and genetic factors. The intracellular aggregation of α-synuclein and the loss of dopaminergic neurons in the substantia nigra pars compacta are the pathological hallmark of PD. At present, there is no successful treatment for PD. Maackiain (MK) is a flavonoid extracted from dried roots of Sophora flavescens Aiton. MK has emerged as a novel agent for PD treatment that acts by inhibiting monoamine oxidase B. In this study, we assessed the neuroprotective potential of MK in Caenorhabditis elegans and investigated possible mechanism of this neuroprotection in the human SH-SY5Y cell line. We found that MK significantly reduced dopaminergic neuron damage in 6-hydroxydopamine (6-OHDA)-exposed worms of the BZ555 strain, with corresponding improvements in food-sensing behavior and life-span. In transgenic worms of strain NL5901 treated with 0.25 mM MK, the accumulation of α-synuclein was diminished by 27% (p < 0.01) compared with that in untreated worms. Moreover, in worms and the SH-SY5Y cell line, we confirmed that the mechanism of MK-mediated protection against PD pathology may include blocking apoptosis, enhancing the ubiquitin-proteasome system, and augmenting autophagy by increasing PINK1/parkin expression. The use of small interfering RNA to downregulate parkin expression in vivo and in vitro could reverse the benefits of MK in PD models. MK may have considerable therapeutic applications in PD.


Asunto(s)
Caenorhabditis elegans/efectos de los fármacos , Neuroblastoma/tratamiento farmacológico , Oxidopamina/toxicidad , Enfermedad de Parkinson/tratamiento farmacológico , Proteínas Quinasas/metabolismo , Pterocarpanos/farmacología , Ubiquitina-Proteína Ligasas/metabolismo , alfa-Sinucleína/toxicidad , Adrenérgicos/toxicidad , Animales , Apoptosis , Autofagia , Caenorhabditis elegans/crecimiento & desarrollo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Neuroblastoma/etiología , Neuroblastoma/patología , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/patología , Proteínas Quinasas/genética , Ubiquitina-Proteína Ligasas/genética
6.
Blood ; 127(10): 1336-45, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26679863

RESUMEN

L5, the most electronegative and atherogenic subfraction of low-density lipoprotein (LDL), induces platelet activation. We hypothesized that plasma L5 levels are increased in acute ischemic stroke patients and examined whether lectin-like oxidized LDL receptor-1 (LOX-1), the receptor for L5 on endothelial cells and platelets, plays a critical role in stroke. Because amyloid ß (Aß) stimulates platelet aggregation, we studied whether L5 and Aß function synergistically to induce prothrombotic pathways leading to stroke. Levels of plasma L5, serum Aß, and platelet LOX-1 expression were significantly higher in acute ischemic stroke patients than in controls without metabolic syndrome (P < .01). In mice subjected to focal cerebral ischemia, L5 treatment resulted in larger infarction volumes than did phosphate-buffered saline treatment. Deficiency or neutralizing of LOX-1 reduced infarct volume up to threefold after focal cerebral ischemia in mice, illustrating the importance of LOX-1 in stroke injury. In human platelets, L5 but not L1 (the least electronegative LDL subfraction) induced Aß release via IκB kinase 2 (IKK2). Furthermore, L5+Aß synergistically induced glycoprotein IIb/IIIa receptor activation; phosphorylation of IKK2, IκBα, p65, and c-Jun N-terminal kinase 1; and platelet aggregation. These effects were blocked by inhibiting IKK2, LOX-1, or nuclear factor-κB (NF-κB). Injecting L5+Aß shortened tail-bleeding time by 50% (n = 12; P < .05 vs L1-injected mice), which was prevented by the IKK2 inhibitor. Our findings suggest that, through LOX-1, atherogenic L5 potentiates Aß-mediated platelet activation, platelet aggregation, and hemostasis via IKK2/NF-κB signaling. L5 elevation may be a risk factor for cerebral atherothrombosis, and downregulating LOX-1 and inhibiting IKK2 may be novel antithrombotic strategies.


Asunto(s)
Isquemia Encefálica/sangre , Lipoproteínas LDL/sangre , Agregación Plaquetaria , Accidente Cerebrovascular/sangre , Péptidos beta-Amiloides/sangre , Animales , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Quinasa I-kappa B/metabolismo , Arteriosclerosis Intracraneal/sangre , Arteriosclerosis Intracraneal/patología , Trombosis Intracraneal/sangre , Trombosis Intracraneal/patología , Masculino , Ratones , Ratones Noqueados , Receptores Depuradores de Clase E/metabolismo , Transducción de Señal , Accidente Cerebrovascular/patología
7.
J Pathol ; 241(3): 337-349, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27801527

RESUMEN

Hypoxia-inducible factor 1α (HIF-1α) controls many genes involved in physiological and pathological processes. However, its roles in glutamatergic transmission and excitotoxicity are unclear. Here, we proposed that HIF-1α might contribute to glutamate-mediated excitotoxicity during cerebral ischaemia-reperfusion (CIR) and investigated its molecular mechanism. We showed that an HIF-1α conditional knockout mouse displayed an inhibition in CIR-induced elevation of extracellular glutamate and N-methyl-d-aspartate receptor (NMDAR) activation. By gene screening for glutamate transporters in cortical cells, we found that HIF-1α mainly regulates the cystine-glutamate transporter (system xc- ) subunit xCT by directly binding to its promoter; xCT and its function are up-regulated in the ischaemic brains of rodents and humans, and the effects lasted for several days. Genetic deletion of xCT in cortical cells of mice inhibits either oxygen glucose deprivation/reoxygenation (OGDR) or CIR-mediated glutamate excitotoxicity in vitro and in vivo. Pharmaceutical inhibition of system xc- by a clinically approved anti-cancer drug, sorafenib, improves infarct volume and functional outcome in rodents with CIR and its therapeutic window is at least 3 days. Taken together, these findings reveal that HIF-1α plays a role in CIR-induced glutamate excitotoxicity via the long-lasting activation of system xc- -dependent glutamate outflow and suggest that system xc- is a promising therapeutic target with an extended therapeutic window in stroke. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/metabolismo , Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Hipoxia de la Célula/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Sistema de Transporte de Aminoácidos y+/genética , Animales , Separación Celular/métodos , Ácido Glutámico/metabolismo , Ratones , Activación Transcripcional/fisiología , Regulación hacia Arriba
8.
Small ; 13(32)2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28665509

RESUMEN

Suspension cells can provide a source of cells for cellular reprogramming, but they are difficult to transfect by nonviral vectors. An efficient and safe nonviral vector (GO-Fe3 O4 -PEI complexes) based on iron oxide nanoparticle (Fe3 O4 )-decorated graphene oxide (GO) complexed with polyethylenimine (PEI) for the first time is developed for delivering three individual episomal plasmids (pCXLE-hOCT3/4-shp53, pCXLE-hSK, and pCXLE-hUL) encoding pluripotent-related factors of Oct3/4, shRNA against p53, Sox2, Klf4, L-Myc, and Lin28 into human peripheral blood mononuclear cells (PBMCs) simultaneously. The combined treatment of magnetic stirring and near-infrared (NIR)-laser irradiation, which can promote contact between the complexes and floating cells and increase the cell membrane permeability, respectively, is used to conduct multiple physical stimulations for suspension PBMCs transfection. The PCR analysis shows that the combinatorial effect of magnetic targeting and photothermal stimulation obviously promoted the transfection efficiency of suspension cells. The transfected cells show positive expression of the pluripotency markers, including Nanog, Oct4, and Sox2, and have potential to differentiate into mesoderm and ectoderm cells. The results demonstrate that the GO-Fe3 O4 -PEI complex provides a safe, convenient, and efficient tool for reprogramming PBMCs into partially induced pluripotent stem cells, which are able to rapidly transdifferentiate into mesodermal lineages without full reprogramming.


Asunto(s)
Linaje de la Célula , Reprogramación Celular , Grafito/farmacología , Magnetismo , Mesodermo/citología , Óxido Ferrosoférrico/química , Humanos , Factor 4 Similar a Kruppel , Polietileneimina/química
9.
Stem Cells ; 33(4): 1153-72, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25523790

RESUMEN

Understanding stem cell homing, which is governed by environmental signals from the surrounding niche, is important for developing effective stem cell-based repair strategies. The molecular mechanism by which the brain under ischemic stress recruits bone marrow-derived cells (BMDCs) to the vascular niche remains poorly characterized. Here we report that hypoxia-inducible factor-1α (HIF-1α) activation upregulates pituitary adenylate cyclase-activating peptide 38 (PACAP38), which in turn activates PACAP type 1 receptor (PAC1) under hypoxia in vitro and cerebral ischemia in vivo. BMDCs homing to endothelial cells in the ischemic brain are mediated by HIF-1α activation of the PACAP38-PAC1 signaling cascade followed by upregulation of cellular prion protein and α6-integrin to enhance the ability of BMDCs to bind laminin in the vascular niche. Exogenous PACAP38 confers a similar effect in facilitating BMDCs homing into the ischemic brain, resulting in reduction of ischemic brain injury. These findings suggest a novel HIF-1α-activated PACAP38-PAC1 signaling process in initiating BMDCs homing into the ischemic brain for reducing brain injury and enhancing functional recovery after ischemic stroke.


Asunto(s)
Células de la Médula Ósea/metabolismo , Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/biosíntesis , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/biosíntesis , Animales , Encéfalo/patología , Células Cultivadas , Células HEK293 , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología
10.
Int J Mol Sci ; 17(12)2016 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-27929425

RESUMEN

To investigate whether the coexistence of hypertension and ovariectomy will increase cardiac Fas receptor and mitochondrial-dependent apoptotic pathways, histopathological analysis, the TUNEL assay and Western blotting were performed on the excised hearts from three groups of female spontaneously hypertensive rats (SHR), which were divided into a sham-operated group (SHR-Sham), bilaterally ovariectomized group (SHR-OVX) and normotensive Wistar Kyoto rats (WKY). Compared with the WKY group, the SHR-Sham group exhibited decreased protein levels of ERα, ERß, p-Akt/Akt, Bcl-2, Bcl-xL and p-Bad and decreased further in the SHR-OVX group, as well as protein levels of t-Bid, Bak, Bad, Bax, cytochrome c, activated caspase-9 and activated caspase-3 (mitochondria-dependent apoptosis) increased in the SHR-Sham group and increased further in the SHR-OVX group. Compared with the WKY group, protein levels of Fas ligand, TNF-α, Fas death receptors, TNFR1, FADD and activated caspase-8 (Fas receptor-dependent apoptosis) increased in the SHR-Sham group, but did not increase in the SHR-OVX group, except Fas ligand and TNF-α. The coexistence of hypertension and ovariectomy attenuated the estrogen receptor survival pathway and appeared to additively increase the cardiac mitochondria-dependent, but not the Fas receptor-dependent apoptosis pathway, which might provide one possible mechanism for the development of cardiac abnormalities in hypertensive postmenopausal women.


Asunto(s)
Apoptosis/fisiología , Hipertensión/complicaciones , Miocardio/patología , Ovariectomía/efectos adversos , Animales , Presión Sanguínea/fisiología , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Femenino , Miocardio/metabolismo , Ratas , Ratas Endogámicas SHR , Ratas Wistar , Factor de Necrosis Tumoral alfa/metabolismo , Receptor fas/metabolismo
11.
FASEB J ; 28(10): 4394-407, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24970391

RESUMEN

Neuronal apoptosis is one of the major causes of poststroke neurological deficits. Inflammation during the acute phase of stroke results in nuclear translocation of NFκB in affected cells in the infarct area. Macrophage migration inhibitory factor (MIF) promotes cardiomyocyte survival in mice following heart ischemia. However, the role of MIF during stroke remains limited. In this study, we showed that MIF expression is down-regulated by 0.75 ± 0.10-fold of the control in the infarct area in the mouse brains. Two functional cis-acing NFκB response elements were identified in the human MIF promoter. Dual activation of hypoxia and NFκB signaling resulted in significant reduction of MIF promoter activity to 0.86 ± 0.01-fold of the control. Furthermore, MIF reduced caspase-3 activation and protected neurons from oxidative stress- and in vitro ischemia/reperfusion-induced apoptosis. H2O2 significantly induced cell death with 12.81 ± 0.58-fold increase of TUNEL-positive cells, and overexpression of MIF blocked the H2O2-induced cell death. Disruption of the MIF gene in MIF-knockout mice resulted in caspase-3 activation, neuronal loss, and increased infarct development during stroke in vivo. The infarct volume was increased from 6.51 ± 0.74% in the wild-type mice to 9.07 ± 0.66% in the MIF-knockout mice. Our study demonstrates that MIF exerts a neuronal protective effect and that down-regulation of MIF by NFκB-mediated signaling under hypoxia accelerates neuronal loss during stroke. Our results suggest that MIF is an important molecule for preserving a longer time window for stroke treatment, and strategies to maintain MIF expression at physiological level could have beneficial effects for stroke patients.


Asunto(s)
Apoptosis , Hipoxia de la Célula , Regulación hacia Abajo , Infarto de la Arteria Cerebral Media/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , FN-kappa B/metabolismo , Neuronas/metabolismo , Animales , Células Cultivadas , Células HEK293 , Humanos , Infarto de la Arteria Cerebral Media/patología , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Masculino , Ratones , Ratones Endogámicos BALB C , FN-kappa B/genética , Estrés Oxidativo
12.
Chin J Physiol ; 58(5): 294-301, 2015 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-26387653

RESUMEN

Hydrocortisone is a growth hormone frequently used in the treatment of low back pain. Hydrocortisone treatment has an anti-inflammation effect, which also inactivates glucose transporter type 4 (GLUT4) by p38 mitogen-activated protein kinase (MAPK) inhibition. Translocation of GLUT4 regulates body glucose homeostasis and muscle repair and is induced by insulin. In this study, 56 SD rats were divided into seven groups, and were treated with insulin or hydrocortisone in sedentary or exercise training groups. The muscle proteins and biochemical blood parameters were analyzed after 7 days of treatments. The results showed that the serum glucose increased in hydrocortisone treatment accompanied by GLUT4 inactivation in both the sedentary and exercise training rats. In the exercise training groups, GLUT4 was redistributed on the plasma membrane on co-treatment with insulin and hydrocortisone through Akt phosphorylation. Insulin treatment exerted a compensatory feedback effect on the GLUT4 translocation on hydrocortisone co-treatment, which was the cause of GLUT4 inactivation.


Asunto(s)
Antiinflamatorios/farmacología , Transportador de Glucosa de Tipo 4/metabolismo , Hidrocortisona/farmacología , Hipoglucemiantes/farmacología , Insulina/farmacología , Músculo Esquelético/efectos de los fármacos , Animales , Antiinflamatorios/uso terapéutico , Interacciones Farmacológicas , Hidrocortisona/uso terapéutico , Hipoglucemiantes/uso terapéutico , Insulina/uso terapéutico , Condicionamiento Físico Animal , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
J Neurosci ; 33(18): 7997-8008, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23637190

RESUMEN

Stroke is the leading cause of disability in developed countries. However, no treatment is available beyond 3 h post-ictus. Here, we report that nuclear translocation of PTEN (phosphatase and tensin homolog deleted on chromosome TEN) is a delayed step causatively leading to excitotoxic (in vitro) and ischemic (in vivo) neuronal injuries. We found that excitotoxic stimulation of N-methyl-d-aspartate (NMDA) resulted in PTEN nuclear translocation in cultured neurons, a process requiring mono-ubiquitination at the lysine 13 residue (K13), as the translocation was prevented by mutation of K13 or a short interfering peptide (Tat-K13) that flanks the K13 residue. More importantly, using a rat model of focal ischemia, we demonstrated that systemic application of Tat-K13, even 6 h after stroke, not only reduced ischemia-induced PTEN nuclear translocation, but also strongly protected against ischemic brain damage. Our study suggests that inhibition of PTEN nuclear translocation may represent a novel after stroke therapy.


Asunto(s)
Isquemia Encefálica/metabolismo , Neuronas/metabolismo , Fosfohidrolasa PTEN/metabolismo , Animales , Encéfalo/citología , Isquemia Encefálica/complicaciones , Isquemia Encefálica/diagnóstico por imagen , Células Cultivadas , Modelos Animales de Enfermedad , Embrión de Mamíferos , Agonistas de Aminoácidos Excitadores/toxicidad , Femenino , Fluorodesoxiglucosa F18 , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Masculino , Ratones , Mutación/genética , N-Metilaspartato/toxicidad , Enfermedades del Sistema Nervioso/etiología , Neuronas/efectos de los fármacos , Señales de Localización Nuclear/genética , Señales de Localización Nuclear/metabolismo , Fosfohidrolasa PTEN/genética , Péptidos/metabolismo , Embarazo , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Cintigrafía , Ratas , Ratas Sprague-Dawley
14.
Adv Sci (Weinh) ; : e2404347, 2024 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-38923327

RESUMEN

Photoimmunotherapy faces challenges due to insufficient intratumoral accumulation of photothermal agents and the reversion of the cancer-immunity cycle during treatment. In this study, an anti-PD-L1-immobilized magnetic gold nanohut, AuNH-2-Ab, with photoresponsive, thermosensitive, and immunomodulatory properties to effectively suppress the growth of primary tumors, elevate immunogenic cell death (ICD) levels, reverse the tumor immune microenvironment (TIME), and consequently inhibit metastases are developed. AuNH-2-Ab achieves high tumor accumulation (9.54% injected dose) following systemic administration, allowing the modulation of hyperthermia dose of over 50 °C in the tumor. By optimizing the hyperthermia dose, AuNH-2-Ab simultaneously target and eliminate cancer cells and tumor-associated macrophages, thereby activating potent antitumor immunity without being compromised by immunosuppressive elements. Hyperthermia/pH induced morphological transformation of AuNH-2-Ab involving the detachment of the surface antibody for in situ PD-L1 inhibition, and exposure of the inner fucoidan layer for natural killer (NK) cell activation. This precision photoimmunotherapy approach reprograms the TIME, significantly prolongs survival in a murine hepatocellular carcinoma model (Hep55.1c), and harnesses the synergistic effects of ICD production and checkpoint inhibitors by utilizing a single nanoplatform.

15.
Neurobiol Dis ; 58: 76-91, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23702312

RESUMEN

Exchange protein activated by cAMP-1 (Epac1) plays an important role in cell proliferation, cell survival and neuronal signaling, and activation of Epac1 in endothelial progenitor cells increases their homing to ischemic muscles and promotes neovascularization in a model of hind limb ischemia. Moreover, upregulation of Epac1 occurs during organ development and in diseases such as myocardial hypertrophy, diabetes, and Alzheimer's disease. We report here that hypoxia upregulated Epac1 through HIF-1α induction in the CD34-immunosorted human umbilical cord blood hematopoietic stem cells (hUCB(34)). Importantly, implantation of hUCB(34) subjected to hypoxia-preconditioning (HP-hUCB(34)) improved stroke outcome, more than did implantation of untreated hUCB(34), in rodents subjected to cerebral ischemia, and this required Epac1-to-matrix metalloprotease (MMP) signaling. This improved therapeutic efficacy correlated with better engraftment and differentiation of these cells in the ischemic host brain. In addition, more than did implantation of untreated HP-hUCB(34), implantation of HP-hUCB(34) improved cerebral blood flow into the ischemic brain via induction of angiogenesis, facilitated proliferation/recruitment of endogenous neural progenitor cells in the ischemic brain, and promoted neurite outgrowth following cerebral ischemia. Consistent with our proposed role of Epac1-to-MMP signaling in hypoxia-preconditioning, the above mentioned effects of implanting HP-hUCB(34) could be abolished by pharmacological inhibition and genetic disruption/deletion of Epac1 or MMPs. We have discovered a HIF-1α-to-Epac1-to-MMP signaling pathway that is required for the improved therapeutic efficacy resulting from hypoxia preconditioning of hUCB(34) in vitro prior to their implantation into the host brain in vivo.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Infarto de la Arteria Cerebral Media , Células Madre Mesenquimatosas/fisiología , Plasticidad Neuronal/fisiología , Regulación hacia Arriba , 2-Metoxiestradiol , Animales , Animales Recién Nacidos , Antígenos CD34/metabolismo , Proliferación Celular , Trasplante de Células Madre de Sangre del Cordón Umbilical , Modelos Animales de Enfermedad , Estradiol/análogos & derivados , Estradiol/farmacología , Glucosa/deficiencia , Proteínas Fluorescentes Verdes/genética , Humanos , Infarto de la Arteria Cerebral Media/diagnóstico por imagen , Infarto de la Arteria Cerebral Media/cirugía , Masculino , Metaloproteinasa 2 de la Matriz/deficiencia , Metaloproteinasa 9 de la Matriz/deficiencia , Ratones Transgénicos , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/genética , Cintigrafía , Ratas , Ratas Sprague-Dawley , Moduladores de Tubulina/farmacología
16.
J Sci Food Agric ; 93(1): 76-84, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22689051

RESUMEN

BACKGROUND: Myricetin is a naturally occurring flavonoid that is found in many fruits, vegetables, teas and medicinal herbs. It has been demonstrated to have anti-inflammatory properties, but, to date, no studies have described the immunomodulatory effects of myricetin on the functions of dendritic cells (DCs). The aim of this study was to evaluate the potential for myricetin to modulate lipopolysaccharide (LPS)-stimulated activation of mouse bone marrow-derived DCs. RESULTS: Our experimental data showed that treatment with myricetin up to 10 µg mL(-1) does not cause cytotoxicity in cells. Myricetin significantly decreased the secretion of tumour necrosis factor-α, interleukin-6 and interleukin-12p70 by LPS-stimulated DCs. The expression of LPS-induced major histocompatibility class II, CD40 and CD86 on DCs was also inhibited by myricetin, and the endocytic and migratory capacity of LPS-stimulated DCs was blocked by myricentin. In addition, LPS-stimulated DC-elicited allogeneic T-cell proliferation was reduced by myricetin. Moreover, our results confirmed that myricetin attenuates the responses of LPS-stimulated activation of DCs via suppression of IκB kinase/nuclear factor-κB and mitogen-activated protein kinase-dependent pathways. CONCLUSION: Myricetin has novel immunopharmacological activity, and modulation of DCs by myricetin may be an attractive strategy for the treatment of inflammatory and autoimmune disorders, and for transplantation.


Asunto(s)
Antiinflamatorios/farmacología , Médula Ósea/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Flavonoides/farmacología , Factores Inmunológicos/farmacología , Inflamación/inmunología , Extractos Vegetales/farmacología , Animales , Antiinflamatorios/uso terapéutico , Antígenos/metabolismo , Médula Ósea/metabolismo , Células Dendríticas/metabolismo , Flavonoides/uso terapéutico , Quinasa I-kappa B/metabolismo , Factores Inmunológicos/uso terapéutico , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Interleucinas/metabolismo , Lipopolisacáridos , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Fitoterapia , Extractos Vegetales/uso terapéutico , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Quinasa de Factor Nuclear kappa B
17.
Biomedicine (Taipei) ; 13(3): 57-62, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37937058

RESUMEN

Background: Recently, immunotherapy has emerged as a promising method for advanced HCC treatment. There are several clinical trials and meta-analyses of immune checkpoint inhibitors and immune cell therapy, but clinical evidence on the combination of these two therapies is lacking. Case description: A 66-year-old man with chronic hepatitis B-related cirrhosis complained of acute abdominal pain in an emergency department of a hospital. On exams, there was a palpable mass in the right upper quadrant of his abdomen. Contrast-enhanced abdominal computed tomography showed a large tumor in the right lobe, 13 cm × 17 cm in size, and right portal vein thrombosis. The alpha-fetoprotein (AFP) level was 30,905 mg/dL. Therefore this patient was diagnosed with BCLC stage C hepatocellular carcinoma (HCC). He underwent trans-arterial chemo-embolization (TACE), abdominal radiotherapy, nivolumab, and lenvatinib. His disease had been under control until two years later, the disease progressed with multiple lung metastases, and his AFP level rose from around 1000 to 17,000 ng/ml. At this stage, he underwent new combination immunotherapy in January 2022. He used pembrolizumab (100 mg) first, and the AFP level decreased by 600 ng/ml daily. Then he received DC-CIK cell therapy two weeks after using pembrolizumab, and the AFP level declined to 900 ng/ml a day. Unfortunately, severe pneumonitis and tension pneumothorax developed after therapy. The patient denied undergoing further treatment and expired peacefully. Conclusion: The previous in-vivo study found that combination immunotherapy can improve tumor control in the mice model. Besides, in previous clinical studies, the level of AFP may be a surrogate marker of tumor response. Therefore we thought the more rapidly declined level of AFP was the clinical evidence of the synergistic effect of checkpoint inhibitors combined with cell therapy in HCC treatment.

18.
Regen Ther ; 24: 443-450, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37753387

RESUMEN

The iPS cells were discovered in 2006. With their ability to differentiate into cells of all three germ layers, iPS cells have great potential for clinical applications. Oct4, Sox2, c-Myc, and Klf4 were identified as the most effective factors for generating iPS cells. Despite this, iPS cells manufactured with these factors would still be inefficient. As a member of the chromobox family, chromobox protein homolog 7 (Cbx7) binds to PRC1 and PRC2 to inhibit genes involved in differentiation. A decrease in the expression of Cbx7 is observed during embryonic stem cell differentiation. Currently, no report discusses the role of Cbx7 in the production of iPS cells. In this study, we hypothesized that Cbx7 could increase iPS cell generation. We confirmed that Cbx7 is highly expressed in pluripotent stem cells (including ES and iPS cells). In addition, transfecting Cbx7 into fibroblasts increased Oct4, Sox2, c-Myc, and Klf4 expression. Moreover, we describe a novel approach to producing iPS cells using Cbx7 in combination with Oct4, Sox2, c-Myc, and Klf4. In summary, we have demonstrated that Cbx7 enhances the reprogramming of iPS cells and characterized the stemness and pluripotency of iPS cells.

19.
Cells ; 12(6)2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36980235

RESUMEN

Glioblastoma (GBM) is a primary brain tumor of unknown etiology. It is extremely aggressive, incurable and has a short average survival time for patients. Therefore, understanding the precise molecular mechanisms of this diseases is essential to establish effective treatments. In this study, we cloned and sequenced a splice variant of the hydroxysteroid 11-ß dehydrogenase 1 like gene (HSD11B1L) and named it HSD11B1L-181. HSD11 B1L-181 was specifically expressed only in GBM cells. Overexpression of this variant can significantly promote the proliferation, migration and invasion of GBM cells. Knockdown of HSD11B1L-181 expression inhibited the oncogenic potential of GBM cells. Furthermore, we identified the direct interaction of parkin with HSD11B1L-181 by screening the GBM cDNA expression library via yeast two-hybrid. Parkin is an RBR E3 ubiquitin ligase whose mutations are associated with tumorigenesis. Small interfering RNA treatment of parkin enhanced the proliferative, migratory and invasive abilities of GBM. Finally, we found that the alkaloid peiminine from the bulbs of Fritillaria thunbergii Miq blocks the interaction between HSD11B1L-181 and parkin, thereby lessening carcinogenesis of GBM. We further confirmed the potential of peiminine to prevent GBM in cellular, ectopic and orthotopic xenograft mouse models. Taken together, these findings not only provide insight into GBM, but also present an opportunity for future GBM treatment.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1 , Neoplasias Encefálicas , Glioblastoma , Ubiquitina-Proteína Ligasas , Animales , Humanos , Ratones , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/efectos de los fármacos , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Carcinogénesis/genética , Cevanas/farmacología , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Isoformas de Proteínas/efectos de los fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ubiquitina-Proteína Ligasas/efectos de los fármacos , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
20.
Nat Commun ; 14(1): 285, 2023 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-36650171

RESUMEN

The potential clinical application of gadolinium-neutron capture therapy (Gd-NCT) for glioblastoma multiforme (GBM) treatment has been compromised by the fast clearance and nonspecific biodistribution of gadolinium-based agents. We have developed a stem cell-nanoparticle system (SNS) to actively target GBM for advanced Gd-NCT by magnetizing umbilical cord mesenchymal stem cells (UMSCs) using gadodiamide-concealed magnetic nanoparticles (Gd-FPFNP). Nanoformulated gadodiamide shielded by a dense surface composed of fucoidan and polyvinyl alcohol demonstrates enhanced cellular association and biocompatibility in UMSCs. The SNS preserves the ability of UMSCs to actively penetrate the blood brain barrier and home to GBM and, when magnetically navigates by an external magnetic field, an 8-fold increase in tumor-to-blood ratio is achieved compared with clinical data. In an orthotopic GBM-bearing rat model, using a single dose of irradiation and an ultra-low gadolinium dose (200 µg kg-1), SNS significantly attenuates GBM progression without inducing safety issues, prolonging median survival 2.5-fold compared to free gadodiamide. The SNS is a cell-based delivery system that integrates the strengths of cell therapy and nanotechnology, which provides an alternative strategy for the treatment of brain diseases.


Asunto(s)
Glioblastoma , Terapia por Captura de Neutrón , Ratas , Animales , Gadolinio , Nanomedicina , Medicina de Precisión , Distribución Tisular , Glioblastoma/tratamiento farmacológico , Neutrones , Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA