Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Analyst ; 145(3): 764-776, 2020 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-31755889

RESUMO

Bone mineral development has been described to proceed through an amorphous precursor prior to apatite crystallization. However, further analytical approaches are necessary to identify specific markers of amorphous mineral components in bone. Here, we establish an original Fourier transform infrared (FTIR) spectroscopy approach to allow the specific identification of the amorphous and/or crystalline nature of bone mineral. Using a series of standards, our results demonstrate that obtaining the second derivative of the FTIR spectra could reveal a peak specifically corresponding to amorphous calcium phosphate (ACP) at ∼992 cm-1. The intensity of this peak was strongly correlated to ACP content in standard mixtures. The analysis of a variety of bones showed that a clear ACP peak could be identified as a specific marker of the existence of an amorphous mineral component in developing bones. In contrast, the ACP peak was not detected in the mature bones. Moreover, subjecting developing bones to ex vivo crystallization conditions led to a clear reduction of the ACP peak, further substantiating the conversion of amorphous mineral precursor into mature apatite crystals. Analysis of mineralization in osteogenic cell cultures corroborated our observations, showing the presence of ACP as a major transient component in early mineralization, but not in the mature matrix. Additionally, FTIR imaging revealed that ACP was present in areas of matrix development, distributed around the edges of mineralizing nodules. Using an original analytical approach, this work provides strong evidence to support that bone mineral development is initiated by an amorphous precursor prior to apatite crystallization.


Assuntos
Osso e Ossos/química , Fosfatos de Cálcio/química , Espectroscopia de Infravermelho com Transformada de Fourier/métodos , Animais , Osso e Ossos/metabolismo , Linhagem Celular , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/química , Osteoblastos/citologia , Osteoblastos/metabolismo , Ratos , Ratos Sprague-Dawley , Peixe-Zebra
2.
Birth Defects Res A Clin Mol Teratol ; 100(6): 453-62, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24954432

RESUMO

BACKGROUND: Myelomeningocele (MMC) is a common congenital malformation and the most severe form of spina bifida characterized by the protrusion of spinal cord and meninges through the spinal defect. Our objective was to improve the assessment of congenital vertebral defects in animal models of MMC using three-dimensional high resolution micro-computed tomography (micro-CT) imaging and quantitative digital analyses methods. METHODS: Lumbosacral MMC was induced in fetal rats by exposure of pregnant mothers at embryonic day 10 (E10) to all-trans retinoic acid, and rats were examined at term (embryonic day 22). The axial skeleton was examined in an MMC model for the first time using ex vivo micro-CT at 10 µm voxel resolution to allow high resolution two-dimensional and three-dimensional characterization of anomalies in lumbosacral vertebrae, and quantitative assessment of distances between dorsal vertebral arches in lumbosacral regions in MMC rats, compared with normal controls. RESULTS: We observed, in detail, skeletal defects in lumbosacral vertebra of MMC rats, including in the morphology of individual dorsal vertebral arches. Use of high resolution micro-CT has also enabled us to identify the delayed (nonfused) or absent ossification in vertebral bodies, increased fusion of adjacent lateral vertebral elements, and quantify the extent of dorsal arch widening. Distances between dorsal vertebral arches showed statistically significant increases from L5 through S4 in MMC rats, compared with normal controls. CONCLUSION: High-resolution micro-CT combined with digital quantification methods is a powerful technique ideally suited for precise assessment of complex congenital skeletal abnormalities such as examined in this rodent model of MMC.


Assuntos
Meningomielocele/patologia , Coluna Vertebral/patologia , Animais , Modelos Animais de Doenças , Feminino , Feto , Humanos , Processamento de Imagem Assistida por Computador , Região Lombossacral , Meningomielocele/induzido quimicamente , Meningomielocele/diagnóstico por imagem , Meningomielocele/embriologia , Gravidez , Ratos , Coluna Vertebral/diagnóstico por imagem , Coluna Vertebral/embriologia , Tretinoína , Microtomografia por Raio-X
3.
Nanomedicine ; 10(8): 1711-8, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24941463

RESUMO

Rebuilding of infarcted myocardium by mesenchymal stem cells (MSCs) has not been successful because of poor cell survival due in part to insufficient blood supply after myocardial infarction (MI). We hypothesize that targeted delivery of vascular endothelial growth factor (VEGF) to MI can help regenerate vasculature in support of MSC therapy in a rat model of MI. VEGF-encapsulated immunoliposomes targeting overexpressed P-selectin in MI tissue were infused by tail vein immediately after MI. One week later, MSCs were injected intramyocardially. The cardiac function loss was moderated slightly by targeted delivery of VEGF or MSC treatment. Targeted VEGF+MSC combination treatment showed highest attenuation in cardiac function loss. The combination treatment also increased blood vessel density (80%) and decreased collagen content in post-MI tissue (33%). Engraftment of MSCs in the combination treatment group was significantly increased and the engrafted cells contributed to the restoration of blood vessels. FROM THE CLINICAL EDITOR: VEGF immunoliposomes targeting myocardial infarction tissue resulted in significantly higher attenuation of cardiac function loss when used in combination with mesenchymal stem cells. MSCs were previously found to have poor ability to restore cardiac tissue, likely as a result of poor blood supply in the affected areas. This new method counterbalances that weakness by the known effects of VEGF, as demonstrated in a rat model.


Assuntos
Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/terapia , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Animais , Vasos Sanguíneos/efeitos dos fármacos , Colágeno/metabolismo , Modelos Animais de Doenças , Transplante de Células-Tronco Mesenquimais , Ratos
4.
Cells ; 12(7)2023 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-37048157

RESUMO

Open neural tube defects (NTDs) such as myelomeningocele (MMC) are debilitating and the most common congenital defects of the central nervous system. Despite their apparent clinical importance, the existing early prenatal diagnostic options for these defects remain limited. Using a well-accepted retinoic-acid-induced model of MMC established in fetal rats, we discovered that neurocan and phosphacan, the secreted chondroitin sulfate proteoglycans of the developing nervous system, are released into the amniotic fluid (AF) of fetal rats displaying spinal cord defects. In contrast to normal controls, elevated AF levels of neurocan and phosphacan were detected in MMC fetuses early in gestation and continued to increase during MMC progression, reaching the highest level in near-term fetuses. The molecular forms of neurocan and phosphacan identified in the AF of MMC fetuses and those found in MMC spinal cords were qualitatively similar. In summary, this is the first report demonstrating the presence of neurocan and phosphacan in the AF of MMC fetuses. The identification of elevated levels of neurocan and phosphacan in the AF of MMC fetuses provides two prospective biomarkers with the potential for early prenatal diagnosis of open NTDs.


Assuntos
Defeitos do Tubo Neural , Neurocam , Gravidez , Feminino , Ratos , Animais , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores , Líquido Amniótico , Biomarcadores , Defeitos do Tubo Neural/diagnóstico
5.
J Cell Physiol ; 227(8): 3119-27, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22034072

RESUMO

An indispensable role for oligodendrocytes in the protection of axon function and promotion of neuronal survival is strongly supported by the finding of progressive neuron/axon degeneration in human neurological diseases that affect oligodendrocytes. Imaging and pathological studies of the CNS have shown the presence of neuroaxonal injury in progressive multifocal leukoencephalopathy (PML), a demyelinating disease of the CNS, resulting from destruction of oligodendrocytes upon productive replication of the pathogenic neurotropic polyomavirus JC. Here, we examined the extracellular factors involved in communication between oligodendrocytes and neurons. Culturing cortical neurons with conditioned medium (CM) from rat CG4 oligodendrocytic cells that express the JCV agnoprotein showed that CXCL5/LIX, which is a chemokine closely related to the human CXCL5/ENA78 and CXCL6/GCP-2 chemokines, is essential for neuronal cell survival. We found that in CM from agnoprotein-producing CG-4 cells level of CXC5/LIX is decreased compared to control cells. We also demonstrated that a reduced expression of CXCL5/LIX by CG4 GFP-Agno cells triggered a cascade of signaling events in cortical neurons. Analysis of mitogen-activated protein kinases (MAPK) and glycogen synthase kinase (GSK3) pathways showed that they are involved in mechanisms of neuronal apoptosis in response to the depletion of CXCL5/LIX signaling. These data suggest that agnoprotein-induced dysregulation of chemokine production by oligodendrocytes may contribute to neuronal/axonal injury in the pathogenesis of PML lesions.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Quimiocina CXCL5/metabolismo , Leucoencefalopatia Multifocal Progressiva/metabolismo , Neurônios/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Meios de Cultivo Condicionados , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Vírus JC/metabolismo , Vírus JC/patogenicidade , Leucoencefalopatia Multifocal Progressiva/patologia , Leucoencefalopatia Multifocal Progressiva/virologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neurônios/citologia , Oligodendroglia/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Proteínas Virais Reguladoras e Acessórias/antagonistas & inibidores
6.
Exp Neurol ; 349: 113961, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34953897

RESUMO

Increasing the intrinsic growth potential of neurons after injury has repeatedly been shown to promote some level of axonal regeneration in rodent models. One of the most studied pathways involves the activation of the PI3K/AKT/mTOR pathways, primarily by reducing the levels of PTEN, a negative regulator of PI3K. Likewise, activation of signal transducer and activator of transcription 3 (STAT3) has previously been shown to boost axonal regeneration and sprouting within the injured nervous system. Here, we examined the regeneration of the corticospinal tract (CST) after cortical expression of constitutively active (ca) Akt3 and STAT3, both separately and in combination. Overexpression of caAkt3 induced regeneration of CST axons past the injury site independent of caSTAT3 overexpression. STAT3 demonstrated improved axon sprouting compared to controls and contributed to a synergistic improvement in effects when combined with Akt3 but failed to promote axonal regeneration as an individual therapy. Despite showing impressive axonal regeneration, animals expressing Akt3 failed to show any functional improvement and deteriorated with time. During this period, we observed progressive Akt3 dose-dependent increase in behavioral seizures. Histology revealed increased phosphorylation of ribosomal S6 protein within the unilateral cortex, increased neuronal size, microglia activation and hemispheric enlargement (hemimegalencephaly).


Assuntos
Axônios , Regeneração Nervosa , Proteínas Proto-Oncogênicas c-akt/biossíntese , Tratos Piramidais/crescimento & desenvolvimento , Tratos Piramidais/lesões , Convulsões/genética , Convulsões/fisiopatologia , Animais , Feminino , Vetores Genéticos , Ativação de Macrófagos , Megalencefalia/patologia , Microglia , Neurônios/patologia , Fosforilação , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Fator de Transcrição STAT3/metabolismo
7.
Exp Cell Res ; 315(3): 462-73, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19061885

RESUMO

Proliferating astrocytic cells from germinal, as well as mature areas of brain parenchyma, have the characteristics of neural stem/progenitor cells and are capable of generating both neurons and glia. We previously reported that primary fetal human brain cells, designated as Normal Human Astrocytes (NHA), expressed, in addition to GFAP, Vimentin and Nestin, low levels of betaIII-Tubulin, an early neuronal marker, and differentiated into neurons and astrocytes in vitro. Here, we showed that primary NHA cells co-express low levels of mesenchymal markers Fibronectin and Collagen-1 in culture. These cells transitioned into mesenchymal-like cells when cultured in adherent conditions in serum containing media. The mesenchymal-like derivatives of these cells were characterized based on their morphological changes, high expression of Vimentin and extracellular matrix (ECM) proteins, Collagen-1 and Fibronectin, and decline of neural markers. When incubated in osteogenic and adipogenic induction media, the mesenchymal-like cells differentiated into osteoblasts and adipocytes. Furthermore, NHA cells express markers of neural crest cells, SOX-10 and p75. These data support the idea of ectoderm-derived mesenchymal lineages. These findings suggest that a population of primitive fetal brain cells with neural/neural crest/mesenchymal phenotype, resembles the remarkable phenotypic plasticity of neural crest cells, and differentiates into adipocytes and osteocytes under the influence of environmental factors.


Assuntos
Astrócitos/citologia , Encéfalo/citologia , Linhagem da Célula , Células-Tronco Mesenquimais/citologia , Adipócitos/citologia , Adipócitos/metabolismo , Antígenos de Diferenciação/metabolismo , Astrócitos/metabolismo , Diferenciação Celular , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Osteoblastos/citologia , Osteoblastos/fisiologia
8.
Front Cell Neurosci ; 14: 201, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32714152

RESUMO

Myelomeningocele (MMC) is the most common congenital defect of the central nervous system and results in devastating and lifelong disability. In MMC, the initial failure of neural tube closure early in gestation is followed by a progressive prenatal injury to the exposed spinal cord, which contributes to the deterioration of neurological function in fetuses. Prenatal strategies to control the spinal cord injury offer an appealing therapeutic approach to improve neurological function, although the definitive pathophysiological mechanisms of injury remain to be fully elucidated. A better understanding of these mechanisms at the cellular and molecular level is of paramount importance for the development of targeted prenatal MMC therapies to minimize or eliminate the effects of the injury and improve neurological function. In this review article, we discuss the pathological development of MMC with a focus on in utero injury to the exposed spinal cord. We emphasize the need for a better understanding of the causative factors in MMC spinal cord injury, pathophysiological alterations associated with the injury, and cellular and molecular mechanisms by which these alterations are induced.

9.
BMC Cancer ; 9: 54, 2009 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-19216795

RESUMO

BACKGROUND: Although features of variable differentiation in glioblastoma cell cultures have been reported, a comparative analysis of differentiation properties of normal neural GFAP positive progenitors, and those shown by glioblastoma cells, has not been performed. METHODS: Following methods were used to compare glioblastoma cells and GFAP+NNP (NHA): exposure to neural differentiation medium, exposure to adipogenic and osteogenic medium, western blot analysis, immunocytochemistry, single cell assay, BrdU incorporation assay. To characterize glioblastoma cells EGFR amplification analysis, LOH/MSI analysis, and P53 nucleotide sequence analysis were performed. RESULTS: In vitro differentiation of cancer cells derived from eight glioblastomas was compared with GFAP-positive normal neural progenitors (GFAP+NNP). Prior to exposure to differentiation medium, both types of cells showed similar multilineage phenotype (CD44+/MAP2+/GFAP+/Vimentin+/Beta III-tubulin+/Fibronectin+) and were positive for SOX-2 and Nestin. In contrast to GFAP+NNP, an efficient differentiation arrest was observed in all cell lines isolated from glioblastomas. Nevertheless, a subpopulation of cells isolated from four glioblastomas differentiated after serum-starvation with varying efficiency into derivatives indistinguishable from the neural derivatives of GFAP+NNP. Moreover, the cells derived from a majority of glioblastomas (7 out of 8), as well as GFAP+NNP, showed features of mesenchymal differentiation when exposed to medium with serum. CONCLUSION: Our results showed that stable co-expression of multilineage markers by glioblastoma cells resulted from differentiation arrest. According to our data up to 95% of glioblastoma cells can present in vitro multilineage phenotype. The mesenchymal differentiation of glioblastoma cells is advanced and similar to mesenchymal differentiation of normal neural progenitors GFAP+NNP.


Assuntos
Glioblastoma/patologia , Neurônios/citologia , Células-Tronco/citologia , Antígeno AC133 , Antígenos CD/biossíntese , Agregação Celular/fisiologia , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Cerebelo/citologia , Receptores ErbB/genética , Genes p53 , Glioblastoma/genética , Glicoproteínas/biossíntese , Humanos , Perda de Heterozigosidade , Mesoderma/patologia , Instabilidade de Microssatélites , Neurônios/patologia , Peptídeos , Células Tumorais Cultivadas
10.
Biomed Microdevices ; 11(5): 1051-7, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19452279

RESUMO

Existing microfluidic devices, e.g. parallel plate flow chambers, do not accurately depict the geometry of microvascular networks in vivo. We have developed a synthetic microvascular network (SMN) on a polydimethalsiloxane (PDMS) chip that can serve as an in vitro model of the bifurcations, tortuosities, and cross-sectional changes found in microvascular networks in vivo. Microvascular networks from a cremaster muscle were mapped using a modified Geographical Information System, and then used to manufacture the SMNs on a PDMS chip. The networks were cultured with bovine aortic endothelial cells (BAEC), which reached confluency 3-4 days after seeding. Propidium iodide staining indicated viable and healthy cells showing normal behavior in these networks. Anti-ICAM-1 conjugated 2-mum microspheres adhered to BAEC cells activated with TNF-alpha in significantly larger numbers compared to control IgG conjugated microspheres. This preferential adhesion suggests that cultured cells retain an intact cytokine response in the SMN. This microfluidic system can provide novel insight into characterization of drug delivery particles and dynamic flow conditions in microvascular networks.


Assuntos
Biomimética/métodos , Vasos Sanguíneos/citologia , Técnicas Analíticas Microfluídicas/métodos , Animais , Bovinos , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Dimetilpolisiloxanos/química , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Humanos , Músculos/irrigação sanguínea , Fator de Necrose Tumoral alfa/farmacologia
11.
J Neurotrauma ; 36(12): 1965-1973, 2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-30284959

RESUMO

Myelomeningocele (MMC) is a devastating congenital neural tube defect that results in the exposure of spinal cord to the intrauterine environment, leading to secondary spinal cord injury and severe impairment. Although the mechanisms underlying the secondary pathogenesis are clinically relevant, the exact cause of in utero-acquired spinal cord damage remains unclear. The objective of this study was to determine whether the hyaluronic acid (HA) concentration in amniotic fluid (AF) in the retinoic acid-induced model of MMC is different from that in normal controls and whether these differences could have an impact on the viscosity of AF. Our data shows that the concentration of HA in AF samples from fetuses with MMC (MMC-AF) and normal control samples (Norm-AF) were not significantly different at embryonic day 18 (E18) and E20. Thereafter, the HA concentration significantly increased in Norm-AF but not in MMC-AF. Compared with Norm-AF, the concentration of HA in MMC-AF and the viscosity of MMC-AF were significantly lower at E21. Agarose gel electrophoresis confirmed a significant reduction in the HA level of MMC-AF compared with Norm-AF at E21. No HA-degrading activity was detected in MMC-AF. In summary, we identified a deficiency in the AF level of HA and the viscosity of AF in fetal rats with MMC. These data are discussed in relation to a potential role the reduction in the AF viscosity due to the low level of HA may play in the exacerbating effects of mechanical trauma on spinal cord damage at the MMC lesion site.


Assuntos
Líquido Amniótico/metabolismo , Ácido Hialurônico/metabolismo , Meningomielocele/metabolismo , Animais , Modelos Animais de Doenças , Meningomielocele/induzido quimicamente , Ratos , Tretinoína
12.
BMC Biotechnol ; 8: 56, 2008 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-18638414

RESUMO

BACKGROUND: Although extensive research has been performed to control differentiation of neural stem cells - still, the response of those cells to diverse cell culture conditions often appears to be random and difficult to predict. To this end, we strived to obtain stabilized protocol of NHA cells differentiation - allowing for an increase in percentage yield of neuronal cells. RESULTS: Uncommitted GFAP and SOX2 positive neural progenitors - so-called, Normal Human Astrocytes (NHA) were differentiated in different environmental conditions to: only neural cells consisted of neuronal [MAP2+, GFAP-] and glial [GFAP+, MAP2-] population, non-neural cells [CD44+, VIMENTIN+, FIBRONECTIN+, MAP2-, GFAP-, S100beta-, SOX2-], or mixture of neural and non-neural cells.In spite of successfully increasing the percentage yield of glial and neuronal vs. non-neural cells by means of environmental changes, we were not able to increase significantly the percentage of neuronal (GABA-ergic and catecholaminergic) over glial cells under several different cell culture testing conditions. Supplementing serum-free medium with several growth factors (SHH, bFGF, GDNF) did not radically change the ratio between neuronal and glial cells--i.e., 1,1:1 in medium without growth factors and 1,4:1 in medium with GDNF, respectively. CONCLUSION: We suggest that biotechnologists attempting to enrich in vitro neural cell cultures in one type of cells - such as that required for transplantology purposes, should consider the strong limiting influence of intrinsic factors upon extracellular factors commonly tested in cell culture conditions.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Neuroglia/citologia , Neuroglia/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/fisiologia , Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Biomarcadores/análise , Biomarcadores/metabolismo , Técnicas de Cultura de Células/métodos , Linhagem da Célula/efeitos dos fármacos , Linhagem da Célula/fisiologia , Células Cultivadas , Meios de Cultura Livres de Soro/farmacologia , Proteínas de Ligação a DNA/análise , Fatores de Crescimento de Fibroblastos/farmacologia , Fibronectinas/análise , Proteína Glial Fibrilar Ácida/análise , Proteínas HMGB/análise , Humanos , Receptores de Hialuronatos/análise , Proteínas Associadas aos Microtúbulos/análise , Fatores de Crescimento Neural/análise , Fatores de Crescimento Neural/farmacologia , Proteínas do Tecido Nervoso/análise , Proteínas do Tecido Nervoso/efeitos dos fármacos , Subunidade beta da Proteína Ligante de Cálcio S100 , Proteínas S100/análise , Fatores de Transcrição SOXB1 , Células-Tronco/citologia , Fatores de Transcrição/análise , Vimentina/análise
13.
Expert Opin Drug Deliv ; 5(4): 459-70, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18426386

RESUMO

BACKGROUND: The development of a number of regenerative strategies in recent years for curing heart disease represents a paradigm shift away from conventional approaches which aim to manage heart disease. Effective administration of pharmaceutical agents targeted directly to the diseased tissue is the key to unlocking the potential of regenerative strategies, which could augment current conventional treatments. OBJECTIVE: The authors review recent advances in targeted drug delivery to diseased cardiac tissue. METHODS: Various therapeutic methodologies designed to selectively deliver pharmaceutical agents to diseased cardiac tissue are discussed in this review. CONCLUSION: Targeted delivery of survival and engraftment promoting factors to damaged cardiac tissue can be an important strategy, for example, in creating a suitable microenvironment encouraging the engraftment of stem cells. Further progress in this emerging field is contingent on the discovery of new biomarkers that are upregulated in damaged cardiac tissue and can be targeted for selective drug delivery. Once fully realized, breakthroughs in this field will have direct applications in the diagnosis and treatment of heart disease through more effective tissue-specific drug delivery and improved imaging modalities.


Assuntos
Fármacos Cardiovasculares/administração & dosagem , Sistemas de Liberação de Medicamentos , Cardiopatias/tratamento farmacológico , Animais , Portadores de Fármacos , Humanos , Miocárdio/patologia , Transplante de Células-Tronco/métodos , Ultrassom
14.
Brain Res ; 1186: 65-73, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17996224

RESUMO

PrP(c) is a cellular isoform of the prion protein with an unknown normal function. One of the putative physiological roles of this protein is its involvement in cell differentiation. Recently, in vitro and in vivo studies showed that GFAP-positive cells have characteristics of stem/progenitor cells that generate neurons and glia. We used an in vitro model of human neurogenesis from GFAP-positive progenitor cells to study the expression of PrP(c) during neural differentiation. Semi-quantitative multiplex-PCR assay and Western blot analysis revealed a significant increase of PRNP expression level in differentiated cells compared to undifferentiated cell population. As determined by immunocytochemistry followed by a quantitative image analysis, the PrP(c) level increased significantly in neuronal cells and did not increase significantly in glial cells. Of note, glial and neuronal cells showed a very large heterogeneity of PrP(c) expression. Our results provide the basis for studying the role of PrP(c) in cell differentiation and neurogenesis from human GFAP-positive progenitor cells.


Assuntos
Astrócitos/metabolismo , Diferenciação Celular/fisiologia , Células-Tronco Fetais/citologia , Neurônios/metabolismo , Proteínas PrPC/metabolismo , Astrócitos/citologia , Linhagem Celular , Cérebro/citologia , Cérebro/metabolismo , Células-Tronco Fetais/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Neurônios/citologia , Proteínas Priônicas , Príons/metabolismo
16.
PLoS One ; 12(3): e0174625, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28358903

RESUMO

Myelomeningocele (MMC) is the most common and severe disabling type of spina bifida resulting in the exposure of vulnerable spinal cord to the hostile intrauterine environment. In this study, we sought to examine the cellular content of fetal amniotic fluid (AF) in MMC and explore a correlation between these cells and pathological development of MMC. MMC was induced in fetal rats by exposing pregnant mothers to all-trans retinoic acid and AF samples were collected before term. Cells were isolated from AF samples and morphologically and phenotypically characterized in short-term cultures. In addition, the spinal cord injury in MMC fetuses was assessed by immunohistochemical examination of astrogliosis. We identified a population of cells from the AF of MMC fetuses (MMC-AF) that formed adherent clusters of tightly packed cells, which were absent from the AF of normal control fetuses (norm-AF). MMC-AF clusters contained cells co-expressing adherens junction associated proteins (ZO-1), N-cadherin and F-actin at sites of cell-cell contacts. In addition, they expressed markers of early neuroepithelial cells such as SOX-1 and Pax-6 along with other stem/progenitor cell markers such as SOX-2 and nestin. Subpopulations of cells in MMC-AF clusters also expressed more advanced differentiation markers such as doublecortin and GFAP. We found that the appearance of cluster forming cells in cultures from MMC-AF correlated with activation of astrogliosis associated with the spinal cord injury in MMC fetuses. In summary, we identified a neuroepithelial cell population in the AF of MMC fetuses that formed adherent clusters in culture and we characterized cellular markers of these cells. Our data suggests that the phase of the disease is a crucial factor in the emergence of these cells into the AF and that these cells may provide a new and important platform for studying the progression of MMC and development of improved strategies for the repair and diagnosis of MMC prenatally.


Assuntos
Líquido Amniótico , Biomarcadores , Meningomielocele/genética , Traumatismos da Medula Espinal/genética , Disrafismo Espinal/metabolismo , Actinas/genética , Animais , Caderinas/genética , Proteína Duplacortina , Feto , Gliose/diagnóstico , Gliose/genética , Gliose/fisiopatologia , Humanos , Meningomielocele/diagnóstico , Meningomielocele/fisiopatologia , Fator de Transcrição PAX6/genética , Diagnóstico Pré-Natal/métodos , Ratos , Medula Espinal/metabolismo , Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/diagnóstico , Traumatismos da Medula Espinal/fisiopatologia , Disrafismo Espinal/diagnóstico , Disrafismo Espinal/genética , Disrafismo Espinal/fisiopatologia , Células-Tronco/metabolismo , Células-Tronco/patologia , Proteína da Zônula de Oclusão-1/genética
17.
PLoS One ; 11(5): e0154726, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27145078

RESUMO

BACKGROUND: The high frequency of mutations in the isocitrate dehydrogenase 1 (IDH1) gene in diffuse gliomas indicates its importance in the process of gliomagenesis. These mutations result in loss of the normal function and acquisition of the neomorphic activity converting α-ketoglutarate to 2-hydroxyglutarate. This potential oncometabolite may induce the epigenetic changes, resulting in the deregulated expression of numerous genes, including those related to the differentiation process or cell survivability. METHODS: Neural stem cells were derived from human induced pluripotent stem cells following embryoid body formation. Neural stem cells transduced with mutant IDH1R132H, empty vector, non-transduced and overexpressing IDH1WT controls were differentiated into astrocytes and neurons in culture. The neuronal and astrocytic differentiation was determined by morphology and expression of lineage specific markers (MAP2, Synapsin I and GFAP) as determined by real-time PCR and immunocytochemical staining. Apoptosis was evaluated by real-time observation of Caspase-3 activation and measurement of PARP cleavage by Western Blot. RESULTS: Compared with control groups, cells expressing IDH1R132H retained an undifferentiated state and lacked morphological changes following stimulated differentiation. The significant inhibitory effect of IDH1R132H on neuronal and astrocytic differentiation was confirmed by immunocytochemical staining for markers of neural stem cells. Additionally, real-time PCR indicated suppressed expression of lineage markers. High percentage of apoptotic cells was detected within IDH1R132H-positive neural stem cells population and their derivatives, if compared to normal neural stem cells and their derivatives. The analysis of PARP and Caspase-3 activity confirmed apoptosis sensitivity in mutant protein-expressing neural cells. CONCLUSIONS: Our study demonstrates that expression of IDH1R132H increases apoptosis susceptibility of neural stem cells and their derivatives. Robust apoptosis causes differentiation deficiency of IDH1R132H-expressing cells.


Assuntos
Apoptose/fisiologia , Diferenciação Celular/fisiologia , Isocitrato Desidrogenase/metabolismo , Células-Tronco Neurais/metabolismo , Astrócitos/metabolismo , Biomarcadores/metabolismo , Caspase 3/metabolismo , Linhagem da Célula/fisiologia , Células Cultivadas , Corpos Embrioides/metabolismo , Glioma/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurogênese/fisiologia , Neurônios/metabolismo
18.
Brain Res ; 1619: 84-90, 2015 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-25498106

RESUMO

Myelomeningocele (MMC) is a devastating spinal cord birth defect, which results in significant life-long disabilities, impaired quality of life, and difficult medical management. The pathological progression of MMC involves failure in neural tube and vertebral arch closure at early gestational ages, followed by subsequent impairment in spinal cord and vertebral growth during fetal development. MMC is irreversible at term. Thus, prenatal therapeutic strategies that interrupt progressive pathological processes offer an appealing approach for treatment of MMC. However, a thorough understanding of pathological progression of MMC is mandatory for appropriate treatment to be rendered. This article is part of a Special Issue entitled SI: Spinal cord injury.


Assuntos
Meningomielocele/patologia , Espinha Bífida Cística/patologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Humanos , Imageamento Tridimensional , Meningomielocele/complicações , Meningomielocele/terapia , Espinha Bífida Cística/complicações , Espinha Bífida Cística/terapia , Microtomografia por Raio-X
19.
PLoS One ; 10(11): e0142725, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26555149

RESUMO

Studies of neonatal neural pathologies and development of appropriate therapeutics are hampered by a lack of relevant in vitro models of neonatal blood-brain barrier (BBB). To establish such a model, we have developed a novel blood-brain barrier on a chip (B3C) that comprises a tissue compartment and vascular channels placed side-by-side mimicking the three-dimensional morphology, size and flow characteristics of microvessels in vivo. Rat brain endothelial cells (RBEC) isolated from neonatal rats were seeded in the vascular channels of B3C and maintained under shear flow conditions, while neonatal rat astrocytes were cultured under static conditions in the tissue compartment of the B3C. RBEC formed continuous endothelial lining with a central lumen along the length of the vascular channels of B3C and exhibited tight junction formation, as measured by the expression of zonula occludens-1 (ZO-1). ZO-1 expression significantly increased with shear flow in the vascular channels and with the presence of astrocyte conditioned medium (ACM) or astrocytes cultured in the tissue compartment. Consistent with in vivo BBB, B3C allowed endfeet-like astrocyte-endothelial cell interactions through a porous interface that separates the tissue compartment containing cultured astrocytes from the cultured RBEC in the vascular channels. The permeability of fluorescent 40 kDa dextran from vascular channel to the tissue compartment significantly decreased when RBEC were cultured in the presence of astrocytes or ACM (from 41.0 ± 0.9 x 10-6 cm/s to 2.9 ± 1.0 x 10-6 cm/s or 1.1±0.4 x 10-6 cm/s, respectively). Measurement of electrical resistance in B3C further supports that the addition of ACM significantly improves the barrier function in neonatal RBEC. Moreover, B3C exhibits significantly improved barrier characteristics compared to the transwell model and B3C permeability was not significantly different from the in vivo BBB permeability in neonatal rats. In summary, we developed a first dynamic in vitro neonatal BBB on a chip (B3C) that closely mimics the in vivo microenvironment, offers the flexibility of real time analysis, and is suitable for studies of BBB function as well as screening of novel therapeutics.


Assuntos
Barreira Hematoencefálica , Dispositivos Lab-On-A-Chip , Animais , Animais Recém-Nascidos , Encéfalo/irrigação sanguínea , Permeabilidade da Membrana Celular , Endotélio Vascular/citologia , Modelos Biológicos , Ratos , Ratos Sprague-Dawley , Proteína da Zônula de Oclusão-1/metabolismo
20.
Methods Mol Biol ; 1078: 89-96, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23975823

RESUMO

Pathological changes in most neurological diseases are marked by cell loss. To understand the mechanisms of neurogenesis and brain repair at a cellular level, observation on less complex systems provide valuable knowledge which offer the basis for therapeutic interventions. This has been the impetus for neural cell culture studies and the development of in vitro models. Here, we provide protocols for differentiation into neuronal lineage of commercially available normal human astrocytes (NHA) that are isolated from normal fetal human brain (Lonza, Inc.). It is known that some of GFAP positive astrocytic cells have stem/progenitor cell characteristics; however, understanding of the human GFAP positive cells with these characteristics remains limited. The genesis of neuronal lineage cells from the NHA occurs in adherent culture conditions by removal of serum and exposure to bFGF. When transferred to serum-free medium supplemented with bFGF, NHA cells generate neuronal precursors that express doublecortin, nestin and are negative for GFAP. After withdrawal of bFGF they mature into neurons. The average time required for generation of neuronal cells using this protocol is about 3 weeks. Our model of neurogenesis captures a contained in vitro system consisting of both neurons and glia. This "human brain in a dish" model can be used to assay the effects of interventions on developing human neurons at a cellular and molecular level and is also suitable for modeling of various aspects of human diseases and testing of novel therapeutic strategies.


Assuntos
Astrócitos/citologia , Diferenciação Celular , Técnicas Citológicas/métodos , Feto/citologia , Neurônios/citologia , Linhagem da Célula , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA