Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Br J Cancer ; 129(5): 797-810, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37474720

RESUMO

BACKGROUND: The aim of this study was to investigate the antitumour activity, safety, and tolerability of pamiparib plus tislelizumab in patients with previously treated advanced solid tumours. METHODS: In this study, patients were enrolled into eight arms by tumour type. All received pamiparib 40 mg orally twice daily plus tislelizumab 200 mg intravenously every 3 weeks. The primary endpoint was objective response rate (ORR), assessed by the investigator per Response Evaluation Criteria in Solid Tumours v1.1. Secondary endpoints included duration of response (DoR), safety, and tolerability. RESULTS: Overall, 180 patients were enrolled. In the overall population, the ORR was 20.0% (range: 0-47.4 across study arms), with median DoR of 17.1 months (95% confidence interval [CI]: 6.2, not estimable [NE]). The highest ORR was observed in the triple-negative breast cancer (TNBC) arm (patients with BRCA1/2 mutations and/or homologous recombination deficiency) (ORR: 47.4%; median DoR: 17.1 months [95% CI: 3.0, NE]). Treatment-emergent adverse events (TEAEs) of ≥Grade 3 occurred in 61.7% of patients. Serious TEAEs occurred in 50.0% of patients. CONCLUSIONS: Pamiparib plus tislelizumab showed a variable level of antitumour activity in patients with advanced solid tumours, with the highest ORR in TNBC and was associated with a manageable safety profile. CLINICAL TRIAL REGISTRATION: ClinicalTrial.gov: NCT02660034.


Assuntos
Proteína BRCA1 , Neoplasias de Mama Triplo Negativas , Humanos , Proteína BRCA2 , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos
2.
Breast Cancer Res Treat ; 197(3): 489-501, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36459284

RESUMO

PURPOSE: To evaluate the efficacy and safety of pamiparib in patients with locally advanced or metastatic human epidermal growth factor receptor 2-negative (HER2-) breast cancer, with deleterious or suspected deleterious germline BRCA1/2 mutations (gBRCA1/2 m). METHODS: In this open-label, phase II, multicenter study in China (NCT03575065), patients with triple-negative breast cancer (TNBC cohort) or hormone receptor-positive (HR+)/HER2- breast cancer (HR+/HER2- cohort) and ≤ 2 prior lines of chemotherapy received pamiparib 60 mg orally twice daily in 28-day, continuous cycles. The primary endpoint was objective response rate (ORR; RECIST v1.1) by independent review committee. RESULTS: In total, 88 patients were enrolled (TNBC cohort: 62; HR+/HER2- cohort: 26). Median age was 45.5 (range: 27-67) years, and 60 patients (68.2%) had received 1 or 2 prior lines of chemotherapy; 42 patients (47.7%) had previously received platinum chemotherapy. In the TNBC cohort, ORR was 38.2% (95% confidence interval [CI] 25.4-52.3) and median duration of response (DoR) was 7.0 months (95% CI 3.9-not estimable). In the HR+/HER2- cohort, ORR was 61.9% (95% CI 38.4-81.9) and median DoR was 7.5 months (95% CI 5.6-14.8). The most common treatment-emergent adverse events (TEAEs), treatment-related TEAEs, and ≥ Grade 3 TEAEs were hematologic (including anemia, decreased neutrophil count, and decreased white blood cell count). Overall, 64.8% of patients had TEAEs leading to dose reduction and 2.3% had TEAEs leading to treatment discontinuation. CONCLUSION: Pamiparib showed encouraging efficacy and an acceptable safety profile in patients with locally advanced and metastatic HER2- breast cancer with gBRCA1/2 m. TRIAL REGISTRATION: ClinicalTrials.gov, NCT03575065; July 2, 2018.


Assuntos
Neoplasias da Mama , Neoplasias de Mama Triplo Negativas , Adulto , Idoso , Feminino , Humanos , Pessoa de Meia-Idade , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Fluorenos/uso terapêutico , Células Germinativas/metabolismo , Mutação , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética
3.
Brain ; 140(3): 692-706, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28115364

RESUMO

Transplanted neurons derived from stem cells have been proposed to improve function in animal models of human disease by various mechanisms such as neuronal replacement. However, whether the grafted neurons receive functional synaptic inputs from the recipient's brain and integrate into host neural circuitry is unknown. Here we studied the synaptic inputs from the host brain to grafted cortical neurons derived from human induced pluripotent stem cells after transplantation into stroke-injured rat cerebral cortex. Using the rabies virus-based trans-synaptic tracing method and immunoelectron microscopy, we demonstrate that the grafted neurons receive direct synaptic inputs from neurons in different host brain areas located in a pattern similar to that of neurons projecting to the corresponding endogenous cortical neurons in the intact brain. Electrophysiological in vivo recordings from the cortical implants show that physiological sensory stimuli, i.e. cutaneous stimulation of nose and paw, can activate or inhibit spontaneous activity in grafted neurons, indicating that at least some of the afferent inputs are functional. In agreement, we find using patch-clamp recordings that a portion of grafted neurons respond to photostimulation of virally transfected, channelrhodopsin-2-expressing thalamo-cortical axons in acute brain slices. The present study demonstrates, for the first time, that the host brain regulates the activity of grafted neurons, providing strong evidence that transplanted human induced pluripotent stem cell-derived cortical neurons can become incorporated into injured cortical circuitry. Our findings support the idea that these neurons could contribute to functional recovery in stroke and other conditions causing neuronal loss in cerebral cortex.


Assuntos
Lesões Encefálicas/cirurgia , Potenciais Somatossensoriais Evocados/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Células-Tronco Pluripotentes Induzidas/transplante , Sinapses/fisiologia , Potenciais de Ação , Vias Aferentes/fisiologia , Animais , Encéfalo/citologia , Encéfalo/ultraestrutura , Lesões Encefálicas/etiologia , Linhagem Celular Transformada , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Córtex Cerebral/ultraestrutura , Modelos Animais de Doenças , Humanos , Lisina/análogos & derivados , Lisina/metabolismo , Masculino , Neurônios/fisiologia , Neurônios/ultraestrutura , Fosfopiruvato Hidratase/metabolismo , Ratos , Ratos Nus , Ratos Sprague-Dawley , Acidente Vascular Cerebral/complicações , Sinapses/ultraestrutura , Núcleos Ventrais do Tálamo/citologia
4.
J Neurosci ; 36(15): 4182-95, 2016 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-27076418

RESUMO

Stroke is a leading cause of disability and currently lacks effective therapy enabling long-term functional recovery. Ischemic brain injury causes local inflammation, which involves both activated resident microglia and infiltrating immune cells, including monocytes. Monocyte-derived macrophages (MDMs) exhibit a high degree of functional plasticity. Here, we determined the role of MDMs in long-term spontaneous functional recovery after middle cerebral artery occlusion in mice. Analyses by flow cytometry and immunocytochemistry revealed that monocytes home to the stroke-injured hemisphere., and that infiltration peaks 3 d after stroke. At day 7, half of the infiltrating MDMs exhibited a bias toward a proinflammatory phenotype and the other half toward an anti-inflammatory phenotype, but during the subsequent 2 weeks, MDMs with an anti-inflammatory phenotype dominated. Blocking monocyte recruitment using the anti-CCR2 antibody MC-21 during the first week after stroke abolished long-term behavioral recovery, as determined in corridor and staircase tests, and drastically decreased tissue expression of anti-inflammatory genes, including TGFß, CD163, and Ym1. Our results show that spontaneously recruited monocytes to the injured brain early after the insult contribute to long-term functional recovery after stroke. SIGNIFICANCE STATEMENT: For decades, any involvement of circulating immune cells in CNS repair was completely denied. Only over the past few years has involvement of monocyte-derived macrophages (MDMs) in CNS repair received appreciation. We show here, for the first time, that MDMs recruited to the injured brain early after ischemic stroke contribute to long-term spontaneous functional recovery through inflammation-resolving activity. Our data raise the possibility that inadequate recruitment of MDMs to the brain after stroke underlies the incomplete functional recovery seen in patients and that boosting homing of MDMs with an anti-inflammatory bias to the injured brain tissue may be a new therapeutic approach to promote long-term improvement after stroke.


Assuntos
Macrófagos , Monócitos , Recuperação de Função Fisiológica , Acidente Vascular Cerebral/fisiopatologia , Animais , Anticorpos Bloqueadores/farmacologia , Antígenos CD/biossíntese , Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/biossíntese , Antígenos de Diferenciação Mielomonocítica/genética , Comportamento Animal/efeitos dos fármacos , Quimera , Lateralidade Funcional , Infarto da Artéria Cerebral Média/fisiopatologia , Inflamação/patologia , Lectinas/biossíntese , Lectinas/genética , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/patologia , Plasticidade Neuronal/fisiologia , Desempenho Psicomotor/efeitos dos fármacos , Receptores CCR2/antagonistas & inibidores , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/genética , Recuperação de Função Fisiológica/efeitos dos fármacos , Acidente Vascular Cerebral/patologia , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/genética , beta-N-Acetil-Hexosaminidases/biossíntese , beta-N-Acetil-Hexosaminidases/genética
5.
J Neuroinflammation ; 14(1): 153, 2017 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-28754163

RESUMO

BACKGROUND: Choroid plexus (CP) supports the entry of monocyte-derived macrophages (MDMs) to the central nervous system in animal models of traumatic brain injury, spinal cord injury, and Alzheimer's disease. Whether the CP is involved in the recruitment of MDMs to the injured brain after ischemic stroke is unknown. METHODS: Adult male C57BL/6 mice were subjected to focal cortical ischemia by permanent occlusion of the distal branch of the right middle cerebral artery. Choroid plexus tissues were collected and analyzed for Vcam1, Madcam1, Cx3cl1, Ccl2, Nt5e, and Ifnγ expression at different timepoints after stroke using qPCR. Changes of MDMs in CP and cerebrospinal fluid (CSF) at 1 day and 3 days after stroke were analyzed using flow cytometry. Infiltration of MDMs into CP and CSF were validated using ß-actin-GFP chimeric mice and Fgd5-CreERT2 x Lox-stop-lox-Tomato mice. CD115+ monocytes were isolated using a magnetic cell separation system from bone marrow of Cx3cr1-GFP or wild-type C57BL/6 donor mice. The freshly isolated monocytes or M2-like MDMs primed in vitro with IL4 and IL13 were stereotaxically injected into the lateral ventricle of stroke-affected mice to trace for their migration into ischemic hemisphere or to assess their effect on post-stroke recovery using open field, corridor, and active avoidance behavioral tests. RESULTS: We found that CP responded to cortical stroke by upregulation of gene expression for several possible mediators of MDM trafficking and, concomitantly, MDMs increased in CP and cerebrospinal fluid (CSF). We then confirmed that MDMs infiltrated from blood into CP and CSF after the insult using ß-actin-GFP chimeric mice and Fgd5-CreERT2 x Lox-stop-lox-Tomato mice. When MDMs were directly administered into CSF following stroke, they homed to the ischemic hemisphere. If they had been primed in vitro prior to their administration to become M2-like macrophages, they promoted post-stroke recovery of motor and cognitive function without influencing infarct volume. CONCLUSIONS: Our findings suggest the possibility that autologous transplantation of M2-like MDMs into CSF might be developed into a new strategy for promoting recovery also in patients with stroke.


Assuntos
Líquido Cefalorraquidiano/metabolismo , Plexo Corióideo/patologia , Macrófagos/patologia , Monócitos/patologia , Acidente Vascular Cerebral/patologia , Actinas/genética , Actinas/metabolismo , Animais , Antígenos CD/metabolismo , Receptor 1 de Quimiocina CX3C/genética , Receptor 1 de Quimiocina CX3C/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Modelos Animais de Doenças , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Monócitos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Acidente Vascular Cerebral/fisiopatologia , Fatores de Tempo , Proteína da Zônula de Oclusão-1/metabolismo
6.
Neurobiol Dis ; 83: 1-15, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26299391

RESUMO

Ischemic stroke triggers neurogenesis from neural stem/progenitor cells (NSPCs) in the subventricular zone (SVZ) and migration of newly formed neuroblasts toward the damaged striatum where they differentiate to mature neurons. Whether it is the injury per se or the associated inflammation that gives rise to this endogenous neurogenic response is unknown. Here we showed that inflammation without corresponding neuronal loss caused by intrastriatal lipopolysaccharide (LPS) injection leads to striatal neurogenesis in rats comparable to that after a 30 min middle cerebral artery occlusion, as characterized by striatal DCX+ neuroblast recruitment and mature NeuN+/BrdU+ neuron formation. Using global gene expression analysis, changes in several factors that could potentially regulate striatal neurogenesis were identified in microglia sorted from SVZ and striatum of LPS-injected and stroke-subjected rats. Among the upregulated factors, one chemokine, CXCL13, was found to promote neuroblast migration from neonatal mouse SVZ explants in vitro. However, neuroblast migration to the striatum was not affected in constitutive CXCL13 receptor CXCR5(-/-) mice subjected to stroke. Infarct volume and pro-inflammatory M1 microglia/macrophage density were increased in CXCR5(-/-) mice, suggesting that microglia-derived CXCL13, acting through CXCR5, might be involved in neuroprotection following stroke. Our findings raise the possibility that the inflammation accompanying an ischemic insult is the major inducer of striatal neurogenesis after stroke.


Assuntos
Corpo Estriado/fisiopatologia , Encefalite/fisiopatologia , Infarto da Artéria Cerebral Média/fisiopatologia , Células-Tronco Neurais/fisiologia , Neurogênese , Neurônios/fisiologia , Acidente Vascular Cerebral/fisiopatologia , Animais , Morte Celular , Movimento Celular/efeitos dos fármacos , Quimiocina CXCL13/farmacologia , Quimiocina CXCL13/fisiologia , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Proteína Duplacortina , Encefalite/induzido quimicamente , Encefalite/metabolismo , Expressão Gênica , Infarto da Artéria Cerebral Média/patologia , Mediadores da Inflamação/metabolismo , Ventrículos Laterais/citologia , Ventrículos Laterais/metabolismo , Ventrículos Laterais/fisiopatologia , Lipopolissacarídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/citologia , Microglia/metabolismo , Neurônios/patologia , Ratos , Ratos Wistar , Receptores CXCR5/genética , Receptores CXCR5/fisiologia , Acidente Vascular Cerebral/patologia
7.
Brain ; 136(Pt 12): 3561-77, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24148272

RESUMO

Stem cell-based approaches to restore function after stroke through replacement of dead neurons require the generation of specific neuronal subtypes. Loss of neurons in the cerebral cortex is a major cause of stroke-induced neurological deficits in adult humans. Reprogramming of adult human somatic cells to induced pluripotent stem cells is a novel approach to produce patient-specific cells for autologous transplantation. Whether such cells can be converted to functional cortical neurons that survive and give rise to behavioural recovery after transplantation in the stroke-injured cerebral cortex is not known. We have generated progenitors in vitro, expressing specific cortical markers and giving rise to functional neurons, from long-term self-renewing neuroepithelial-like stem cells, produced from adult human fibroblast-derived induced pluripotent stem cells. At 2 months after transplantation into the stroke-damaged rat cortex, the cortically fated cells showed less proliferation and more efficient conversion to mature neurons with morphological and immunohistochemical characteristics of a cortical phenotype and higher axonal projection density as compared with non-fated cells. Pyramidal morphology and localization of the cells expressing the cortex-specific marker TBR1 in a certain layered pattern provided further evidence supporting the cortical phenotype of the fated, grafted cells, and electrophysiological recordings demonstrated their functionality. Both fated and non-fated cell-transplanted groups showed bilateral recovery of the impaired function in the stepping test compared with vehicle-injected animals. The behavioural improvement at this early time point was most likely not due to neuronal replacement and reconstruction of circuitry. At 5 months after stroke in immunocompromised rats, there was no tumour formation and the grafted cells exhibited electrophysiological properties of mature neurons with evidence of integration in host circuitry. Our findings show, for the first time, that human skin-derived induced pluripotent stem cells can be differentiated to cortical neuronal progenitors, which survive, differentiate to functional neurons and improve neurological outcome after intracortical implantation in a rat stroke model.


Assuntos
Córtex Cerebral/citologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Infarto da Artéria Cerebral Média/cirurgia , Neurônios/fisiologia , Recuperação de Função Fisiológica/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/transplante , Modelos Animais de Doenças , Estimulação Elétrica , Glutaminase/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Infarto da Artéria Cerebral Média/patologia , Neurônios/classificação , Neurônios/efeitos dos fármacos , Neurotransmissores/farmacologia , Técnicas de Patch-Clamp , Ratos , Ratos Nus , Ratos Sprague-Dawley
8.
PLoS One ; 13(2): e0192118, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29401502

RESUMO

Induced pluripotent stem cells (iPSCs) or their progeny, derived from human somatic cells, can give rise to functional improvements after intracerebral transplantation in animal models of stroke. Previous studies have indicated that reactive gliosis, which is associated with stroke, inhibits neurogenesis from both endogenous and grafted neural stem/progenitor cells (NSPCs) of rodent origin. Here we have assessed whether reactive astrocytes affect the fate of human iPSC-derived NSPCs transplanted into stroke-injured brain. Mice with genetically attenuated reactive gliosis (deficient for GFAP and vimentin) were subjected to cortical stroke and cells were implanted adjacent to the ischemic lesion one week later. At 8 weeks after transplantation, immunohistochemical analysis showed that attenuated reactive gliosis did not affect neurogenesis or commitment towards glial lineage of the grafted NSPCs. Our findings, obtained in a human-to-mouse xenograft experiment, provide evidence that the reactive gliosis in stroke-injured brain does not affect the formation of new neurons from intracortically grafted human iPSC-derived NSPCs. However, for a potential clinical translation of these cells in stroke, it will be important to clarify whether the lack of effect of reactive gliosis on neurogenesis is observed also in a human-to-human experimental setting.


Assuntos
Gliose/prevenção & controle , Células-Tronco Pluripotentes Induzidas/transplante , Células-Tronco Neurais/transplante , Neurogênese , Acidente Vascular Cerebral/patologia , Animais , Proteína Glial Fibrilar Ácida/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação
9.
Exp Neurol ; 297: 129-137, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28746827

RESUMO

Ischemic stroke, caused by middle cerebral artery occlusion, leads to long-lasting formation of new striatal neurons from neural stem/progenitor cells (NSPCs) in the subventricular zone (SVZ) of adult rodents. Concomitantly with this neurogenic response, SVZ exhibits activation of resident microglia and infiltrating monocytes. Here we show that depletion of circulating monocytes, using the anti-CCR2 antibody MC-21 during the first week after stroke, enhances striatal neurogenesis at one week post-insult, most likely by increasing short-term survival of the newly formed neuroblasts in the SVZ and adjacent striatum. Blocking monocyte recruitment did not alter the volume of the ischemic lesion but gave rise to reduced astrocyte activation in SVZ and adjacent striatum, which could contribute to the improved neuroblast survival. A similar decrease of astrocyte activation was found in and around human induced pluripotent stem cell (iPSC)-derived NSPCs transplanted into striatum at one week after stroke in monocyte-depleted mice. However, there was no effect on neurogenesis in the graft as determined 8weeks after implantation. Our findings demonstrate, for the first time, that a specific cellular component of the early inflammatory reaction in SVZ and adjacent striatum following stroke, i.e., infiltrating monocytes, compromises the short-term neurogenic response neurogenesis from endogenous NSPCs.


Assuntos
Encéfalo/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Monócitos/fisiologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Acidente Vascular Cerebral/terapia , Fatores Etários , Animais , Encéfalo/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/transplante , Transplante de Células-Tronco/métodos , Acidente Vascular Cerebral/patologia
10.
Stem Cell Res Ther ; 8(1): 59, 2017 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-28279192

RESUMO

BACKGROUND: Intracerebral transplantation of human induced pluripotent stem cells (iPSCs) can ameliorate behavioral deficits in animal models of stroke. How the ischemic lesion affects the survival of the transplanted cells, their proliferation, migration, differentiation, and function is only partly understood. METHODS: Here we have assessed the influence of the stroke-induced injury on grafts of human skin iPSCs-derived long-term neuroepithelial-like stem cells using transplantation into the rostral migratory stream (RMS), adjacent to the neurogenic subventricular zone, in adult rats as a model system. RESULTS: We show that the occurrence of an ischemic lesion, induced by middle cerebral artery occlusion, in the striatum close to the transplant does not alter the survival, proliferation, or generation of neuroblasts or mature neurons or astrocytes from the grafted progenitors. In contrast, the migration and axonal projection patterns of the transplanted cells are markedly influenced. In the intact brain, the grafted cells send many fibers to the main olfactory bulb through the RMS and a few of them migrate in the same direction, reaching the first one third of this pathway. In the stroke-injured brain, on the other hand, the grafted cells only migrate toward the ischemic lesion and virtually no axonal outgrowth is observed in the RMS. CONCLUSIONS: Our findings indicate that signals released from the stroke-injured area regulate the migration of and fiber outgrowth from grafted human skin-derived neural progenitors and overcome the influence on these cellular properties exerted by the neurogenic area/RMS in the intact brain.


Assuntos
Células-Tronco Pluripotentes Induzidas/transplante , Células-Tronco Neurais/transplante , Neurogênese , Acidente Vascular Cerebral/terapia , Animais , Astrócitos/metabolismo , Axônios/metabolismo , Encéfalo/patologia , Diferenciação Celular/genética , Humanos , Infarto da Artéria Cerebral Média , Células-Tronco Neurais/imunologia , Neurônios/imunologia , Neurônios/patologia , Ratos , Acidente Vascular Cerebral/imunologia , Acidente Vascular Cerebral/patologia
11.
Int J Biol Sci ; 12(5): 505-17, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27019633

RESUMO

Directed differentiation of human induced pluripotent stem cells (iPSCs) into retinal pigmented epithelium (RPE) holds great promise in cell replacement therapy for patients suffering from degenerative eye diseases, including age-related macular degeneration (AMD). In this study, we generated iPSCs from human dermal fibroblasts (HDFs) by electroporation with episomal plasmid vectors encoding OCT4, SOX2, KLF4, L-MYC together with p53 suppression. Intriguingly, cell reprogramming resulted in a metastable transcriptional activation and selective demethylation of neural and retinal specification-associated genes, such as OTX2, RX1 and SIX3. In contrast, RPE progenitor genes were transcriptionally silent in HDFs and descendant iPSCs. Overexpression of OCT4 and SOX2 directly stimulated the expression of OTX2, RX1 and SIX3 in HDFs and iPSCs. Luciferase and chromatin immunoprecipitation (ChIP) assays further identified an OCT4- and two SOX2-binding sites located in the proximal promoter of OTX2. Histone acetylation and methylation on the local promoter also participated in the reactivation of OTX2. The transcriptional conversion of RX1 and SIX3 genes partially attributed to DNA demethylation. Subsequently, iPSCs were induced into the RPE cells displaying the characteristics of polygonal shapes and pigments, and expressing typical RPE cell markers. Taken together, our results establish readily efficient and safe protocols to produce iPSCs and iPSC-derived RPE cells, and underline that the reactivation of anterior neural transcription factor OTX2, eye field transcription factor RX1 and SIX3 in iPSCs is a feature of pluripotency acquisition and predetermines the potential of RPE differentiation.


Assuntos
Proteínas do Olho/genética , Proteínas de Homeodomínio/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas do Tecido Nervoso/genética , Fator 3 de Transcrição de Octâmero/genética , Fatores de Transcrição Otx/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Reprogramação Celular/genética , Reprogramação Celular/fisiologia , Imunoprecipitação da Cromatina , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Proteínas Proto-Oncogênicas c-myc/genética , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição SOXB1/genética , Proteína Homeobox SIX3
12.
Int J Biol Sci ; 7(6): 892-901, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21814484

RESUMO

In this report, we describe the spontaneous malignant transformation of long-term cultured human fetal striatum neural stem cells (hsNSCs, passage 17). After subcutaneous transplantation of long-term cultured hsNSCs into immunodeficient nude mice, 2 out of 15 mice formed xenografts which expressed neuroendocrine tumor markers CgA and NSE. T1 cells, a cell line that we derived from one of the two subcutaneous xenografts, have undergone continuous expansion in vitro. These T1 cells showed stem cell-like features and expressed neural stem cell markers nestin and CD133. The T1 cells were involved in abnormal karyotype, genomic instability and fast proliferation. Importantly, after long-term in vitro culture, the T1 cells did not result in subcutaneous xenografts, but induced intracranial tumor formation, indicating that they adjusted themselves to the intracranial microenvironment. We further found that the T1 cells exhibited an overexpressed level of EGFR, and the CD133 positive T1 cells showed a truncation mutation in the exons 2-7 of the EGFR (EGFRvIII) gene. These results suggest that continuous expansion of neural stem cells in culture may lead to malignant spontaneous transformation. This phenomenon may be functionally related to EGFR by EGFRvIII gene mutation.


Assuntos
Transformação Celular Neoplásica , Células-Tronco Neurais/fisiologia , Animais , Neoplasias Encefálicas/etiologia , Receptores ErbB/genética , Amplificação de Genes , Genes erbB-1 , Células HeLa , Humanos , Imunofenotipagem , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Experimentais , Neoplasias de Tecido Nervoso/etiologia , Células-Tronco Neurais/patologia
13.
Neurosci Lett ; 484(3): 210-4, 2010 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-20800650

RESUMO

An increasing number of studies support the presence of stem-like cells in human malignancies. These cells are primarily responsible for tumor initiation and thus considered as a potential target to eradicate tumors. CD133 has been identified as an important cell surface marker to enrich the stem-like population in various human tumors. However, the biological function of CD133 protein remains unknown. In this study, we observed no significant effects on cell proliferation and migration in CD133 overexpressed U87MG human glioblastoma cells. It is reported that MAPK/Erk was constitutively activated in CD133 positive liver cancer stem cell. To find out possible mechanism between CD133 and Erk phosphorylation, we performed this study to evaluate the level of Erk phosphorylation in CD133 overexpressed U87MG cells. We found that CD133 overexpression significantly activated Erk, which suggested CD133 involved in activation of MAPK/Erk pathway.


Assuntos
Antígenos CD/genética , Neoplasias Encefálicas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glioblastoma/metabolismo , Glicoproteínas/genética , Peptídeos/genética , Antígeno AC133 , Antígenos CD/biossíntese , Antígenos CD/fisiologia , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/patologia , Carcinoma Hepatocelular/enzimologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Ativação Enzimática/genética , Glioblastoma/enzimologia , Glioblastoma/patologia , Glicoproteínas/biossíntese , Glicoproteínas/fisiologia , Células HEK293 , Humanos , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Sistema de Sinalização das MAP Quinases/genética , Peptídeos/fisiologia , Fosforilação/genética , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA