Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Stroke ; 47(4): 1094-100, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26965847

RESUMO

BACKGROUND AND PURPOSE: Pentraxin 3 (PTX3) is released on inflammatory responses in many organs. However, roles of PTX3 in brain are still mostly unknown. Here we asked whether and how PTX3 contributes to blood-brain barrier dysfunction during the acute phase of ischemic stroke. METHODS: In vivo, spontaneously hypertensive rats were subjected to focal cerebral ischemia by transient middle cerebral artery occlusion. At day 3, brains were analyzed to evaluate the cellular origin of PTX3 expression. Correlations with blood-brain barrier breakdown were assessed by IgG staining. In vitro, rat primary astrocytes and rat brain endothelial RBE.4 cells were cultured to study the role of astrocyte-derived PTX3 on vascular endothelial growth factor-mediated endothelial permeability. RESULTS: During the acute phase of stroke, reactive astrocytes in the peri-infarct area expressed PTX3. There was negative correlation between gradients of IgG leakage and PTX3-positive astrocytes. Cell culture experiments showed that astrocyte-conditioned media increased levels of tight junction proteins and reduced endothelial permeability under normal conditions. Removing PTX3 from astrocyte-conditioned media by immunoprecipitation increased endothelial permeability. PTX3 strongly bound vascular endothelial growth factor in vitro and was able to decrease vascular endothelial growth factor-induced endothelial permeability. CONCLUSIONS: Astrocytes in peri-infarct areas upregulate PTX3, which may support blood-brain barrier integrity by regulating vascular endothelial growth factor-related mechanisms. This response in astrocytes may comprise a compensatory mechanism for maintaining blood-brain barrier function after ischemic stroke.


Assuntos
Astrócitos/metabolismo , Barreira Hematoencefálica/metabolismo , Proteína C-Reativa/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Componente Amiloide P Sérico/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Barreira Hematoencefálica/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Morte Celular , Meios de Cultivo Condicionados , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Infarto da Artéria Cerebral Média/patologia , Masculino , Ratos , Ratos Endogâmicos SHR , Acidente Vascular Cerebral/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
J Biol Chem ; 285(22): 17209-17, 2010 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20212047

RESUMO

Stem cell biology offers advantages to investigators seeking to identify new therapeutic molecules. Specifically, stem cells are genetically stable, scalable for molecular screening, and function in cellular assays for drug efficacy and safety. A key hurdle for drug discoverers of central nervous system disease is a lack of high quality neuronal cells. In the central nervous system, alpha-amino-3-hydroxyl-5-methyl-4-isoxazolepropionate (AMPA) subtype glutamate receptors mediate the vast majority of excitatory neurotransmissions. Embryonic stem (ES) cell protocols were developed to differentiate into neuronal subtypes that express AMPA receptors and were pharmacologically responsive to standard compounds for AMPA potentiation. Therefore, we hypothesized that stem cell-derived neurons should be predictive in high-throughput screens (HTSs). Here, we describe a murine ES cell-based HTS of a 2.4 x 10(6) compound library, the identification of novel chemical "hits" for AMPA potentiation, structure function relationship of compounds and receptors, and validation of chemical leads in secondary assays using human ES cell-derived neurons. This reporting of murine ES cell derivatives being formatted to deliver HTS of greater than 10(6) compounds for a specific drug target conclusively demonstrates a new application for stem cells in drug discovery. In the future new molecular entities may be screened directly in human ES or induced pluripotent stem cell derivatives.


Assuntos
Células-Tronco Embrionárias/citologia , Neurônios/metabolismo , Receptores de AMPA/química , Receptores de Glutamato/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia , Animais , Química Farmacêutica/métodos , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Fluorometria/métodos , Humanos , Imuno-Histoquímica/métodos , Camundongos , Modelos Biológicos , Mutação , Tecnologia Farmacêutica/métodos
3.
Curr Opin Pharmacol ; 7(5): 515-20, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17681491

RESUMO

All physiologic processes operate in a cellular setting. Therefore, drug discoverers need the highest quality cells as they pursue the next generation of safe and effective medicines. Recently, investigators have begun to consider stem cells as a new source of predictive, cell-based assays in drug discovery. Stem cell technology still has hurdles to overcome before these cells are fully accepted as decision-making reagents and amenable to high-throughput screening. However, with global research interest in stem cell biology, significant advances in the application of these cells in drug discovery have been reported. These advances are aligned with three important stages of pharmaceutical research: target discovery and validation, identification of efficacious chemical leads, and drug safety pharmacology. This concise review describes the application of stem cells in these areas of drug discovery with emphasis on molecular screening opportunities.


Assuntos
Desenho de Fármacos , Células-Tronco , Animais , Diferenciação Celular , Humanos , Células-Tronco/citologia
4.
J Clin Invest ; 112(2): 197-208, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12843127

RESUMO

The serine/threonine kinase Akt/PKB plays key roles in the regulation of cell growth, survival, and metabolism. It remains unclear, however, whether the functions of individual Akt/PKB isoforms are distinct. To investigate the function of Akt2/PKBbeta, mice lacking this isoform were generated. Both male and female Akt2/PKBbeta-null mice exhibit mild growth deficiency and an age-dependent loss of adipose tissue or lipoatrophy, with all observed adipose depots dramatically reduced by 22 weeks of age. Akt2/PKBbeta-deficient mice are insulin resistant with elevated plasma triglycerides. In addition, Akt2/PKBbeta-deficient mice exhibit fed and fasting hyperglycemia, hyperinsulinemia, glucose intolerance, and impaired muscle glucose uptake. In males, insulin resistance progresses to a severe form of diabetes accompanied by pancreatic beta cell failure. In contrast, female Akt2/PKBbeta-deficient mice remain mildly hyperglycemic and hyperinsulinemic until at least one year of age. Thus, Akt2/PKBbeta-deficient mice exhibit growth deficiency similar to that reported previously for mice lacking Akt1/PKBalpha, indicating that both Akt2/PKBbeta and Akt1/PKBalpha participate in the regulation of growth. The marked hyperglycemia and loss of pancreatic beta cells and adipose tissue in Akt2/PKBbeta-deficient mice suggest that Akt2/PKBbeta plays critical roles in glucose metabolism and the development or maintenance of proper adipose tissue and islet mass for which other Akt/PKB isoforms are unable to fully compensate.


Assuntos
Tecido Adiposo/patologia , Envelhecimento , Diabetes Mellitus Experimental/patologia , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/fisiologia , Tecido Adiposo/metabolismo , Animais , Peso Corporal , Caspase 3 , Caspases/metabolismo , Feminino , Vetores Genéticos , Glucose/metabolismo , Teste de Tolerância a Glucose , Glucose-6-Fosfatase/metabolismo , Glicogênio Sintase/metabolismo , Hiperglicemia/genética , Hiperglicemia/patologia , Hiperinsulinismo/genética , Imuno-Histoquímica , Insulina/sangue , Insulina/metabolismo , Ilhotas Pancreáticas/patologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Modelos Genéticos , Músculos/metabolismo , Tamanho do Órgão , Fenótipo , Fosfoenolpiruvato Carboxiquinase (GTP)/biossíntese , Fosfoenolpiruvato Carboxiquinase (GTP)/genética , Reação em Cadeia da Polimerase , Isoformas de Proteínas , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt , Fatores de Tempo , Tomografia Computadorizada por Raios X
5.
Mol Cell Biol ; 22(9): 3035-45, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11940661

RESUMO

While scaffold proteins are thought to be key components of signaling pathways, their exact function is unknown. By preassembling multiple components of signaling cascades, scaffolds are predicted to influence the efficiency and/or specificity of signaling events. Here we analyze a potential scaffold of the Ras/mitogen-activated protein kinase (MAPK) pathway, kinase suppressor of Ras (KSR), by generating KSR-deficient mice. KSR-deficient mice were grossly normal even though ERK kinase activation was attenuated to a degree sufficient to block T-cell activation and inhibit tumor development. Consistent with its role as a scaffold, high-molecular-weight complexes containing KSR, MEK, and ERK were lost in the absence of KSR. This demonstrates that KSR is a bona fide scaffold that is not required for but enhances signaling via the Ras/MAPK signaling pathway.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Quinases/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Divisão Celular , Ativação Enzimática , Fibroblastos , Citometria de Fluxo , Deleção de Genes , Marcação de Genes , Interferon gama/análise , Interleucina-2/metabolismo , Interleucina-4/análise , Ativação Linfocitária , Linfócitos/citologia , Linfócitos/imunologia , Linfócitos/metabolismo , MAP Quinase Quinase 1 , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Knockout , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Peso Molecular , Fenótipo , Proteínas Quinases/deficiência , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Células Tumorais Cultivadas
6.
Neuropharmacology ; 51(2): 374-85, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16769090

RESUMO

PDE10A is a newly identified phosphodiesterase that is highly expressed by the medium spiny projection neurons of the striatum. In order to investigate the physiological role of PDE10A in the central nervous system, PDE10A knockout mice (PDE10A(-/-)) were characterized both behaviorally and neurochemically. PDE10A(-/-) mice showed decreased exploratory activity and a significant delay in the acquisition of conditioned avoidance behavior when compared to wild-type (PDE10A(+/+)) mice. However, in a variety of other well-characterized behavioral tasks, including the elevated plus maze (anxiety), forced swim test (depression), hot plate (nociception) and two memory models (passive avoidance and Morris water maze), PDE10A(-/-) mice performed similarly to wild-type mice. When challenged with PCP or MK-801, PDE10A(-/-) mice showed a blunted locomotor response in comparison to PDE10A(+/+) mice. In contrast, PDE10A(-/-) and PDE10A(+/+) mice responded similarly to the locomotor stimulating effects of amphetamine and methamphetamine. Our findings suggest that PDE10A is involved in regulating striatal output, possibly by reducing the sensitivity of medium spiny neurons to glutamatergic excitation. These results are discussed in relationship to the hypothesis that PDE10A inhibition presents a novel treatment for psychosis.


Assuntos
Corpo Estriado/fisiologia , Diester Fosfórico Hidrolases/fisiologia , Anfetamina/farmacologia , Animais , Aprendizagem da Esquiva , Comportamento Animal , Monoaminas Biogênicas/metabolismo , Encéfalo/metabolismo , Estimulantes do Sistema Nervoso Central/farmacologia , Condicionamento Operante , Corpo Estriado/enzimologia , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Aprendizagem em Labirinto , Metanfetamina/farmacologia , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Fenciclidina/farmacologia , Diester Fosfórico Hidrolases/genética
7.
Toxicol Sci ; 90(1): 149-58, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16357009

RESUMO

Embryonic stem (ES) cells offer unprecedented opportunities for in vitro drug discovery and safety assessment of compounds. Cardiomyocytes derived from ES cells enable development of predictive cardiotoxicity models to increase the safety of novel drugs. Heterogeneity of differentiated ES cells limits the development of reliable in vitro models for compound screening. We report an innovative and robust approach to isolate ES-derived cardiomyocytes using laser microdissection and pressure catapulting (LMPC). LMPC cells were readily applied onto 96-well format in vitro pharmacology assays. The expression of developmental and functional cardiac markers, Nkx 2.5, MLC2V, GATA-4, Connexin 43, Connexin 45, Serca-2a, cardiac alpha actin, and phospholamban, among others, was confirmed in LMPC ES-derived cardiomyocytes. Functional assays exhibited cardiac-like response to increased extracellular calcium (5.4 mM extracellular Ca2+) and L-type calcium channel antagonist (1 microM nifedipine). In conclusion, laser microdissection and pressure catapulting is a robust technology to isolate homogeneous ES-derived cell types from heterogeneous populations applicable to assay development.


Assuntos
Cardiopatias/induzido quimicamente , Microscopia Confocal/métodos , Miócitos Cardíacos/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Xenobióticos/toxicidade , Animais , Bioensaio/métodos , Biomarcadores/metabolismo , Cálcio/metabolismo , Cálcio/farmacologia , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos/métodos , Coração Fetal/citologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Cardiopatias/patologia , Lasers , Camundongos , Camundongos Endogâmicos DBA , Microdissecção/métodos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Nifedipino/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos , Células-Tronco/metabolismo , Células-Tronco/patologia
8.
J Biol Rhythms ; 20(2): 122-31, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15834109

RESUMO

In vitro neuronal recordings in the SCN have clearly documented shifts in the peak of unit activity following the application of serotonergic agents, and yet selectivity issues with these very tools have limited progress in establishing the precise receptor mechanisms. As an alternative strategy, mice were bred (C57BL/6J) lacking 1 serotonin receptor, the 5-HT(7), to serve as a null background for this subtype; earlier work had documented the involvement of 5-HT(7) receptors in the phase advances elicited by 8-OH-DPAT, a mixed 5-HT(1A/7) agonist, in SCN slices prepared from rat donors. Single-unit recordings in sequential electrode passes revealed peaks of activity that occurred at nearly the same time in the knockout (KO; ZT4.2 +/- 0.6) and wild-type animals (WT; ZT4.3 +/- 0.1), where ZT0 marks the beginning of the light phase in a 12:12 LD cycle. Bath application of 8-OH-DPAT produced a phase advance in neuronal firing (2.1 +/- 0.5 h) when applied 1 circadian cycle earlier at ZT6 (10 microM, 10 min), but surprisingly, the mean phase advance in slices prepared from KO mice (2.3 +/- 0.1 h) was no different. Coapplication of 8-OH-DPAT with WAY-100,635 (10 microM), a highly selective 5-HT(1A) antagonist, significantly reduced the phase advance, both in experiments with WT and KO mice, suggesting the greater importance of this serotonin sub-type independent of genetic modification. 5-HT itself (0.5 +/-M, 10 min) at ZT6 also yielded phase advances that were indistinguishable in slices prepared from WT and KO mice (1.8 +/- 0.4 h and 2.1 +/- 0.2 h, respectively) and that were also sensitive to WAY-100,635. Unlike the pattern with 8-OH-DPAT, however, 5-HT-induced phase advances, in both WT and KO mice, were blocked by ritanserin, in this paradigm useful as a 5-HT(5A/7) antagonist (in addition to its more typical role as a 5-HT2A/2C antagonist). Serotonin antagonists when administered alone were without effect in slices from WT mice but produced significant phase shifts when administered to those from KO animals. Taken together, these results highlight the importance of the species used in establishing receptor mechanism. More provocatively, they support the involvement of multiple serotonin receptors in shifting the phase of circadian rhythms at ZT6.


Assuntos
8-Hidroxi-2-(di-n-propilamino)tetralina/farmacologia , Ritmo Circadiano , Neurônios/efeitos dos fármacos , Receptores de Serotonina/fisiologia , Serotonina/farmacologia , Núcleo Supraquiasmático/efeitos dos fármacos , Animais , Técnicas In Vitro , Camundongos , Camundongos Knockout , Neurônios/fisiologia , Fenótipo , Receptores de Serotonina/genética , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/fisiologia
9.
J Pharmacol Toxicol Methods ; 81: 240-50, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27184445

RESUMO

INTRODUCTION: The Comprehensive in vitro Proarrhythmia Assay (CiPA) initiative seeks an in vitro test to accurately predict clinical Torsades de Pointes (TdP). We developed a cardiotoxicity assay incorporating simultaneous measurement of the action potential (AP) waveform and Ca(2+) transient (CT) in human iPSC-derived cardiomyocytes (CMs). Concurrent optogenetic pacing provided a well-controlled electrophysiological background. METHODS: We used the Optopatch platform for all-optical electrophysiology (Hochbaum et al., 2014). In a monolayer culture, a subset of cells expressed a genetically encoded, calcium and voltage reporter, CaViar (Hou, Kralj, Douglass, Engert, & Cohen, 2014), while others expressed a channelrhodopsin variant, CheRiff. Optical pacing of CheRiff-expressing cells synchronized the syncytium. We screened 12 compounds (11 acute, 1 chronic) to identify electrophysiological (AP rise time, AP50, AP90, beat rate) and CT effects in spontaneously beating and paced cultures (1Hz, 2Hz). RESULTS: CaViar reported spontaneous and paced APs and CTs with high signal-to-noise ratio and low phototoxicity. Quinidine, flecainide, E-4031, digoxin and cisapride prolonged APs, while verapamil and nifedipine shortened APs. Early after depolarizations (EADs) were elicited by quinidine, flecainide and cisapride. All but four compounds (amiodarone, chromanol, nifedipine, verapamil) prolonged AP rise time. Nifedipine and verapamil decreased CT amplitude, while digoxin increased CT amplitude. Pentamidine prolonged APs after chronic exposure. DISCUSSION: The Optopatch platform provides a robust assay to measure APs and CTs in hiPSC-CMs. This addresses the CiPA mandate and will facilitate comparisons of cell-based assays to human clinical data.


Assuntos
Cardiotoxicidade , Imagem Molecular/métodos , Optogenética/métodos , Potenciais de Ação/efeitos dos fármacos , Arritmias Cardíacas/induzido quimicamente , Arritmias Cardíacas/fisiopatologia , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Estimulação Cardíaca Artificial , Avaliação Pré-Clínica de Medicamentos/métodos , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Humanos , Miócitos Cardíacos/efeitos dos fármacos , Razão Sinal-Ruído
10.
Cell Transplant ; 25(4): 705-14, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26811151

RESUMO

Aging and vascular comorbidities such as hypertension comprise critical cofactors that influence how the brain responds to stroke. Ischemic stress induces neurogenesis and oligodendrogenesis in younger brains. However, it remains unclear whether these compensatory mechanisms can be maintained even under pathologically hypertensive and aged states. To clarify the age-related remodeling capacity after stroke under hypertensive conditions, we assessed infarct volume, behavioral outcomes, and surrogate markers of neurogenesis and oligodendrogenesis in acute and subacute phases after transient focal cerebral ischemia in 3- and 12-month-old spontaneously hypertensive rats (SHRs). Hematoxylin and eosin staining showed that 3- and 12-month-old SHRs exhibited similar infarction volumes at both 3 and 14 days after focal cerebral ischemia. However, recovery of behavioral deficits (neurological score assessment and adhesive removal test) was significantly less in 12-month-old SHRs compared to 3-month-old SHRs. Concomitantly, numbers of nestin(+) neural stem/progenitor cells (NSPCs) near the infarct border area or subventricular zone in 12-month-old SHRs were lower than 3-month-old SHRs at day 3. Similarly, numbers of PDGFR-α(+) oligodendrocyte precursor cells (OPCs) in the corpus callosum were lower in 12-month-old SHRs at day 3. Lower levels of NSPC and OPC numbers were accompanied by lower expression levels of phosphorylated CREB. By day 14 postischemia, NSPC and OPC numbers in 12-month-old SHRs recovered to similar levels as in 3-month-old SHRs, but the numbers of proliferating NSPCs (Ki-67(+)nestin(+) cells) and proliferating OPCs (Ki-67(+)PDGFR-α(+) cells) remained lower in the older brains even at day 14. Taken together, these findings suggest that aging may also decrease poststroke compensatory responses for neurogenesis and oligodendrogenesis even under hypertensive conditions.


Assuntos
Envelhecimento/metabolismo , Infarto Encefálico/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese , Oligodendroglia/metabolismo , Envelhecimento/patologia , Animais , Infarto Encefálico/patologia , Masculino , Células-Tronco Neurais/patologia , Oligodendroglia/patologia , Ratos , Ratos Endogâmicos SHR
12.
PLoS One ; 10(9): e0138724, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26407349

RESUMO

Adult mammalian brain can be plastic after injury and disease. Therefore, boosting endogenous repair mechanisms would be a useful therapeutic approach for neurological disorders. Isoxazole-9 (Isx-9) has been reported to enhance neurogenesis from neural stem/progenitor cells (NSPCs). However, the effects of Isx-9 on other types of progenitor/precursor cells remain mostly unknown. In this study, we investigated the effects of Isx-9 on the three major populations of progenitor/precursor cells in brain: NSPCs, oligodendrocyte precursor cells (OPCs), and endothelial progenitor cells (EPCs). Cultured primary NSPCs, OPCs, or EPCs were treated with various concentrations of Isx-9 (6.25, 12.5, 25, 50 µM), and their cell numbers were counted in a blinded manner. Isx-9 slightly increased the number of NSPCs and effectively induced neuronal differentiation of NSPCs. However, Isx-9 significantly decreased OPC number in a concentration-dependent manner, suggesting cytotoxicity. Isx-9 did not affect EPC cell number. But in a matrigel assay of angiogenesis, Isx-9 significantly inhibited tube formation in outgrowth endothelial cells derived from EPCs. This potential anti-tube-formation effect of Isx-9 was confirmed in a brain endothelial cell line. Taken together, our data suggest that mechanisms and targets for promoting stem/progenitor cells in the central nervous system may significantly differ between cell types.


Assuntos
Diferenciação Celular , Células Progenitoras Endoteliais/efeitos dos fármacos , Isoxazóis/farmacologia , Células-Tronco Neurais/efeitos dos fármacos , Animais , Biomarcadores , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Células Progenitoras Endoteliais/citologia , Células Progenitoras Endoteliais/metabolismo , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese , Neurônios/citologia , Neurônios/metabolismo , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Ratos
13.
PLoS One ; 8(2): e56024, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23418498

RESUMO

We have used a previously unavailable model of pancreatic development, derived in vitro from human embryonic stem cells, to capture a time-course of gene, miRNA and histone modification levels in pancreatic endocrine cells. We investigated whether it is possible to better understand, and hence control, the biological pathways leading to pancreatic endocrine formation by analysing this information and combining it with the available scientific literature to generate models using a casual reasoning approach. We show that the embryonic stem cell differentiation protocol is highly reproducible in producing endocrine precursor cells and generates cells that recapitulate many aspects of human embryonic pancreas development, including maturation into functional endocrine cells when transplanted into recipient animals. The availability of whole genome gene and miRNA expression data from the early stages of human pancreatic development will be of great benefit to those in the fields of developmental biology and diabetes research. Our causal reasoning algorithm suggested the involvement of novel gene networks, such as NEUROG3/E2F1/KDM5B and SOCS3/STAT3/IL-6, in endocrine cell development We experimentally investigated the role of the top-ranked prediction by showing that addition of exogenous IL-6 could affect the expression of the endocrine progenitor genes NEUROG3 and NKX2.2.


Assuntos
Diferenciação Celular/genética , Linhagem da Célula/genética , Redes Reguladoras de Genes , Ilhotas Pancreáticas/metabolismo , Algoritmos , Animais , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Teste de Tolerância a Glucose , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Ilhotas Pancreáticas/embriologia , Camundongos , Proteínas Nucleares , Fatores de Transcrição
15.
PLoS One ; 6(6): e20692, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21673956

RESUMO

The availability of human neuronal progenitors (hNPs) in high purity would greatly facilitate neuronal drug discovery and developmental studies, as well as cell replacement strategies for neurodegenerative diseases and conditions, such as spinal cord injury, stroke, Parkinson's disease, Alzheimer's disease, and Huntington's disease. Here we describe for the first time a method for producing hNPs in large quantity and high purity from human embryonic stem cells (hESCs) in feeder-free conditions, without the use of exogenous noggin, sonic hedgehog or analogs, rendering the process clinically compliant. The resulting population displays characteristic neuronal-specific markers. When allowed to spontaneously differentiate into neuronal subtypes in vitro, cholinergic, serotonergic, dopaminergic and/or noradrenergic, and medium spiny striatal neurons were observed. When transplanted into the injured spinal cord the hNPs survived, integrated into host tissue, and matured into a variety of neuronal subtypes. Our method of deriving neuronal progenitors from hESCs renders the process amenable to therapeutic and commercial use.


Assuntos
Técnicas Citológicas/métodos , Células-Tronco Embrionárias/citologia , Neurônios/citologia , Animais , Diferenciação Celular , Meios de Cultura Livres de Soro , Feminino , Humanos , Ratos
16.
Regen Med ; 5(4): 557-68, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20632859

RESUMO

Pluripotent embryonic stem cells (ESCs), when compared with transformed, primary or engineered cells, have unique characteristics and advantages that have resulted in the development of important cell-based tools in modern drug discovery. However, a key limitation has been the availability of human ESCs from patients with specific medical needs and the broad range of genetic variation represented worldwide. Induced pluripotent stem (iPS) cells are derived from somatic cells that are reprogrammed to a pluripotent stem cell state and have functional characteristics similar to ESCs. The demonstration that human iPS cells can be derived, with relative ease, through the introduction of transcription factor combinations has allowed the generation of disease-specific iPS cell lines. Therefore, iPS cell technology may deliver robust, human pluripotent cell lines from a wide range of clinical phenotypes and genotypes. Although human iPS cell technology is still a new tool in drug discovery, the promise that this technology will impact the discovery of new therapies can be projected based on the uptake of stem cell applications in biopharmaceutical sciences. Here, the near-term opportunities that iPS cells may deliver to drug discoverers to generate and test hypotheses will be discussed, with a focus on the specific strengths and weaknesses of iPS cell technology. Finally, the future perspective will address novel opportunities iPS cells could uniquely deliver to the preclinical development of new drug therapies.


Assuntos
Pesquisa Biomédica , Diferenciação Celular/efeitos dos fármacos , Descoberta de Drogas/história , Ensaios de Triagem em Larga Escala/métodos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Animais , História do Século XXI , Humanos , Células-Tronco Pluripotentes Induzidas/citologia
17.
Cell Stem Cell ; 6(6): 517-20, 2010 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-20569688

RESUMO

Human stem cell biology is driving the promise of novel regenerative therapies into clinical trials. Although the pharmaceutical industry has embraced stem cells as tools in drug discovery, few companies have taken the risk to deliver stem cell-based medicines. Here, we evaluate the various cell-based opportunities and corporate strategies.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Ensaios Clínicos como Assunto , Descoberta de Drogas , Células-Tronco , Terapia Baseada em Transplante de Células e Tecidos/economia , Análise Custo-Benefício , Atenção à Saúde , Indústria Farmacêutica/economia , Humanos
18.
Methods Mol Biol ; 602: 37-54, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20012391

RESUMO

Genetically modified mouse models have been proven to be a powerful tool in drug discovery. The ability to genetically modify the mouse genome by removing or replacing a specific gene has enhanced our ability to identify and validate target genes of interest. In addition, many human diseases can be mimicked in the mouse and signaling pathways have been shown to be conserved. In spite of these advantages the technology has limitations. In transgenic animals there may be significant heterogeneity among different founders. In knock-out animals the predicted phenotypes are not always readily observed and occasionally a completely novel and unexpected phenotype emerges. To address the latter and ensure that a deep knowledge of the target of interest is obtained, we have developed a comprehensive phenotyping program which has identified novel phenotypes as well as any potential safety concerns which may be associated with a particular target. Finally we continue to explore innovative technologies as they become available such as RNAi for temporal and spatial gene knock-down and humanized models that may better simulate human disease states.


Assuntos
Descoberta de Drogas , Indústria Farmacêutica , Camundongos Transgênicos , Animais , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Humanos , Camundongos , Camundongos Knockout , Fenótipo
19.
Cell Metab ; 10(5): 366-78, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19883615

RESUMO

Kinase suppressors of Ras 1 and 2 (KSR1 and KSR2) function as molecular scaffolds to potently regulate the MAP kinases ERK1/2 and affect multiple cell fates. Here we show that KSR2 interacts with and modulates the activity of AMPK. KSR2 regulates AMPK-dependent glucose uptake and fatty acid oxidation in mouse embryonic fibroblasts and glycolysis in a neuronal cell line. Disruption of KSR2 in vivo impairs AMPK-regulated processes affecting fatty acid oxidation and thermogenesis to cause obesity. Despite their increased adiposity, ksr2(-/-) mice are hypophagic and hyperactive but expend less energy than wild-type mice. In addition, hyperinsulinemic-euglycemic clamp studies reveal that ksr2(-/-) mice are profoundly insulin resistant. The expression of genes mediating oxidative phosphorylation is also downregulated in the adipose tissue of ksr2(-/-) mice. These data demonstrate that ksr2(-/-) mice are highly efficient in conserving energy, revealing a novel role for KSR2 in AMPK-mediated regulation of energy metabolism.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Metabolismo Energético , Resistência à Insulina , Proteínas Serina-Treonina Quinases/metabolismo , Tecido Adiposo/metabolismo , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Metabolismo Energético/fisiologia , Ácidos Graxos/metabolismo , Glucose/metabolismo , Glicólise/fisiologia , Resistência à Insulina/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Knockout , Obesidade/etiologia , Obesidade/metabolismo , Oxirredução , Fosforilação Oxidativa , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Termogênese/fisiologia
20.
J Immunol ; 176(5): 3141-8, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16493073

RESUMO

We previously described the in vitro characteristics of the potent and selective CCR1 antagonist, CP-481,715. In addition to being selective for CCR1 vs other chemokine receptors, CP-481,715 is also specific for human CCR1 (hCCR1), preventing its evaluation in classical animal models. To address this, we generated mice whereby murine CCR1 was replaced by hCCR1 (knockin) and used these animals to assess the anti-inflammatory properties of CP-481,715. Cells isolated from hCCR1 knockin mice were shown to express hCCR1 and migrate in response to both murine CCR1 and hCCR1 ligands. Furthermore, this migration is inhibited by CP-481,715 at dose levels comparable to those obtained with human cells. In animal models of cell infiltration, CP-481,715 inhibited CCL3-induced neutrophil infiltration into skin or into an air pouch with an ED50 of 0.2 mg/kg. CP-481,715 did not inhibit cell infiltration in wild-type animals expressing murine CCR1. In a more generalized model of inflammation, delayed-type hypersensitivity, CP-481,715 significantly inhibited footpad swelling and decreased the amount of IFN-gamma and IL-2 produced by isolated spleen cells from sensitized animals. It did not, however, induce tolerance to a subsequent challenge. These studies illustrate the utility of hCCR1 knockin animals to assess the activity of human specific CCR1 antagonists; demonstrate the ability of the CCR1 antagonist CP-481,715 to inhibit cell infiltration, inflammation, and Th1 cytokine responses in these animals; and suggest that CP-481,715 may be useful to modulate inflammatory responses in human disease.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Inibição de Migração Celular , Quimiotaxia de Leucócito/efeitos dos fármacos , Hipersensibilidade Tardia/patologia , Quinoxalinas/farmacologia , Receptores de Quimiocinas/antagonistas & inibidores , Receptores de Quimiocinas/genética , Actinas/metabolismo , Animais , Células Cultivadas , Quimiocina CCL3 , Quimiocina CCL4 , Quimiocinas CC/fisiologia , Quimiotaxia de Leucócito/imunologia , Citocinas/metabolismo , Humanos , Hipersensibilidade Tardia/tratamento farmacológico , Hipersensibilidade Tardia/genética , Proteínas Inflamatórias de Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Receptores CCR1 , Células-Tronco/imunologia , Células-Tronco/patologia , Células Th1/efeitos dos fármacos , Células Th1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA