Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Fluids Barriers CNS ; 15(1): 15, 2018 May 15.
Article in English | MEDLINE | ID: mdl-29759080

ABSTRACT

Zika virus (ZIKV) is a flavivirus that is highly neurotropic causing congenital abnormalities and neurological damage to the central nervous systems (CNS). In this study, we used a human induced pluripotent stem cell (iPSC)-derived blood brain barrier (BBB) model to demonstrate that ZIKV can infect brain endothelial cells (i-BECs) without compromising the BBB barrier integrity or permeability. Although no disruption to the BBB was observed post-infection, ZIKV particles were released on the abluminal side of the BBB model and infected underlying iPSC-derived neural progenitor cells (i-NPs). AXL, a putative ZIKV cellular entry receptor, was also highly expressed in ZIKV-susceptible i-BEC and i-NPs. This iPSC-derived BBB model can help elucidate the mechanism by which ZIKV can infect BECs, cross the BBB and gain access to the CNS.


Subject(s)
Blood-Brain Barrier/metabolism , Blood-Brain Barrier/virology , Zika Virus/metabolism , Capillary Permeability/physiology , Cell Culture Techniques , Endothelial Cells/metabolism , Endothelial Cells/virology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/virology , Microvessels/metabolism , Microvessels/virology , Zika Virus Infection/metabolism , Zika Virus Infection/virology
2.
Sci Rep ; 8(1): 1873, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29382846

ABSTRACT

We have developed a renewable, scalable and transgene free human blood-brain barrier model, composed of brain endothelial cells (BECs), generated from human amniotic fluid derived induced pluripotent stem cells (AF-iPSC), which can also give rise to syngeneic neural cells of the neurovascular unit. These AF-iPSC-derived BECs (i-BEC) exhibited high transendothelial electrical resistance (up to 1500 Ω cm2) inducible by astrocyte-derived molecular cues and retinoic acid treatment, polarized expression of functional efflux transporters and receptor mediated transcytosis triggered by antibodies against specific receptors. In vitro human BBB models enable pre-clinical screening of central nervous system (CNS)-targeting drugs and are of particular importance for assessing species-specific/selective transport mechanisms. This i-BEC human BBB model discriminates species-selective antibody- mediated transcytosis mechanisms, is predictive of in vivo CNS exposure of rodent cross-reactive antibodies and can be implemented into pre-clinical CNS drug discovery and development processes.


Subject(s)
Antibodies/pharmacology , Blood-Brain Barrier/metabolism , Brain/metabolism , Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Receptors, Cell Surface/metabolism , Transcytosis/physiology , Animals , Astrocytes/cytology , Astrocytes/physiology , Biological Transport , Blood-Brain Barrier/drug effects , Brain/drug effects , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/physiology , Humans , Induced Pluripotent Stem Cells/physiology , Male , Neurons/cytology , Neurons/physiology , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/antagonists & inhibitors
3.
Biochem Cell Biol ; 96(4): 391-406, 2018 08.
Article in English | MEDLINE | ID: mdl-29370536

ABSTRACT

Traumatic brain injury (TBI) is a leading cause of morbidity and mortality worldwide. Due to its high incidence rate and often long-term sequelae, TBI contributes significantly to increasing costs of health care expenditures annually. Unfortunately, advances in the field have been stifled by patient and injury heterogeneity that pose a major challenge in TBI prevention, diagnosis, and treatment. In this review, we briefly discuss the causes of TBI, followed by its prevalence, classification, and pathophysiology. The current imaging detection methods and animal models used to study brain injury are examined. We discuss the potential use of molecular markers in detecting and monitoring the progression of TBI, with particular emphasis on microRNAs as a novel class of molecular modulators of injury and its repair in the neural tissue.


Subject(s)
Biomarkers/analysis , Brain Injuries, Traumatic , Functional Neuroimaging , MicroRNAs/therapeutic use , Animals , Brain/diagnostic imaging , Brain Injuries, Traumatic/diagnosis , Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/therapy , Disease Models, Animal , Humans
4.
Crit Care Med ; 44(9): e846-53, 2016 09.
Article in English | MEDLINE | ID: mdl-27071071

ABSTRACT

OBJECTIVE: Diabetic ketoacidosis in children is associated with vasogenic cerebral edema, possibly due to the release of destructive polymorphonuclear neutrophil azurophilic enzymes. Our objectives were to measure plasma azurophilic enzyme levels in children with diabetic ketoacidosis, to correlate plasma azurophilic enzyme levels with diabetic ketoacidosis severity, and to determine whether azurophilic enzymes disrupt the blood-brain barrier in vitro. DESIGN: Prospective clinical and laboratory study. SETTING: The Children's Hospital, London Health Sciences Centre. SUBJECTS: Pediatric type 1 diabetes patients; acute diabetic ketoacidosis or age-/sex-matched insulin-controlled. MEASUREMENTS AND MAIN RESULTS: Acute diabetic ketoacidosis in children was associated with elevated polymorphonuclear neutrophils. Plasma azurophilic enzymes were elevated in diabetic ketoacidosis patients, including human leukocyte elastase (p < 0.001), proteinase-3 (p < 0.01), and myeloperoxidase (p < 0.001). A leukocyte origin of human leukocyte elastase and proteinase-3 in diabetic ketoacidosis was confirmed with buffy coat quantitative real-time polymerase chain reaction (p < 0.01). Of the three azurophilic enzymes elevated, only proteinase-3 levels correlated with diabetic ketoacidosis severity (p = 0.002). Recombinant proteinase-3 applied to human brain microvascular endothelial cells degraded both the tight junction protein occludin (p < 0.05) and the adherens junction protein VE-cadherin (p < 0.05). Permeability of human brain microvascular endothelial cell monolayers was increased by recombinant proteinase-3 application (p = 0.010). CONCLUSIONS: Our results indicate that diabetic ketoacidosis is associated with systemic polymorphonuclear neutrophil activation and degranulation. Of all the polymorphonuclear neutrophil azurophilic enzymes examined, only proteinase-3 correlated with diabetic ketoacidosis severity and potently degraded the blood-brain barrier in vitro. Proteinase-3 might mediate vasogenic edema during diabetic ketoacidosis, and selective proteinase-3 antagonists may offer future vascular- and neuroprotection.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Edema/enzymology , Diabetic Ketoacidosis/enzymology , Leukocyte Elastase/blood , Myeloblastin/blood , Peroxidase/blood , Brain Edema/etiology , Case-Control Studies , Cathepsin G/blood , Cell Culture Techniques , Child , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/enzymology , Diabetic Ketoacidosis/complications , Endothelial Cells/physiology , Female , Humans , Male
5.
Expert Opin Drug Discov ; 10(2): 141-55, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25388782

ABSTRACT

INTRODUCTION: The majority of therapeutics, small molecule or biologics, developed for the CNS do not penetrate the blood-brain barrier (BBB) sufficiently to induce pharmacologically meaningful effects on CNS targets. To improve the efficiency of CNS drug discovery, several in vitro models of the BBB have been used to aid early selection of molecules with CNS exposure potential. However, correlative studies suggest relatively poor predictability of in vitro BBB models underscoring the need to combine in vitro and in vivo BBB penetration assessment into an integrated preclinical workflow. AREAS COVERED: This review gives a brief general overview of in vitro and in vivo BBB models used in the pre-clinical evaluation of CNS-targeting drugs, with particular focus on the recent progress in developing humanized models. The authors discuss the advantages, limitations, in vitro-in vivo correlation, and integration of these models into CNS drug discovery and development with the aim of improving translation. EXPERT OPINION: Often, a simplistic rationalization of the CNS drug discovery and development process overlooks or even ignores the need for an early and predictive assessment of the BBB permeability. Indeed, past failures of CNS candidates in clinical trials argue strongly that the early deployment of in vitro and in vivo models for assessing BBB permeability, mechanisms of transport and brain exposure of leads, and the co-development of BBB delivery strategies will improve translation and increase the clinical success of CNS pipelines.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Drug Discovery/methods , Models, Biological , Animals , Antibodies, Monoclonal/pharmacology , Blood-Brain Barrier/cytology , Brain/cytology , Brain/drug effects , Brain/metabolism , Drug Evaluation, Preclinical , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Species Specificity
6.
Can J Physiol Pharmacol ; 92(12): 1001-11, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25388371

ABSTRACT

S-nitrosoglutathione (GSNO) is an endogenously produced S-nitrosylating compound that controls the function of various proteins. While a number of rodent cell lines have been used to study GSNO-induced apoptosis, the mechanisms of action remain to be evaluated in human cells and in parallel with other common apoptosis-inducing agents. In this study, we compared the pro-apoptotic effects of GSNO and staurosporine (STS) on human neural progenitors (NT2, hNP1) and neuroblasts (SH-SY5Y). We show that these cells exhibit comparable levels of susceptibility to GSNO- and STS-induced apoptotic cell death, as demonstrated by condensed nuclei and CASP3 activation. Mechanistic differences in apoptotic responses were observed as differential patterns of DNA fragmentation and levels of BAX, BCL-XL, CASP8, and p-ERK in response to GSNO and STS treatment. Mitochondrial membrane potential analysis revealed that NT2 and hNP1 cells, but not SH-SY5Y cells, undergo mitochondrial hyperpolarization in response to short-term exposure to STS prior to undergoing subsequent depolarization. This is the first study to report differences in apoptotic responses to GSNO and STS in 3 complementary human neural cell lines. Furthermore, these cells represent useful tools in cell pharmacological paradigms in which susceptibility to apoptosis-inducing agents needs to be assessed at different stages of neural cell fate commitment and differentiation.


Subject(s)
Apoptosis/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , S-Nitrosoglutathione/pharmacology , Staurosporine/pharmacology , Apoptosis Regulatory Proteins/metabolism , Cell Differentiation , Cell Line , Cell Nucleus/pathology , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , S-Nitrosoglutathione/metabolism
7.
Stem Cell Rev Rep ; 10(2): 251-68, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24415130

ABSTRACT

Brain injury continues to be one of the leading causes of disability worldwide. Despite decades of research, there is currently no pharmacologically effective treatment for preventing neuronal loss and repairing the brain. As a result, novel therapeutic approaches, such as cell-based therapies, are being actively pursued to repair tissue damage and restore neurological function after injury. In this study, we examined the neuroprotective potential of amniotic fluid (AF) single cell clones, engineered to secrete glial cell derived neurotrophic factor (AF-GDNF), both in vitro and in a surgically induced model of brain injury. Our results show that pre-treatment with GDNF significantly increases cell survival in cultures of AF cells or cortical neurons exposed to hydrogen peroxide. Since improving the efficacy of cell transplantation depends on enhanced graft cell survival, we investigated whether AF-GDNF cells seeded on polyglycolic acid (PGA) scaffolds could enhance graft survival following implantation into the lesion cavity. Encouragingly, the AF-GDNF cells survived longer than control AF cells in serum-free conditions and continued to secrete GDNF both in vitro and following implantation into the injured motor cortex. AF-GDNF implantation in the acute period following injury was sufficient to activate the MAPK/ERK signaling pathway in host neural cells in the peri-lesion area, potentially boosting endogenous neuroprotective pathways. These results were complemented with promising trends in beam walk tasks in AF-GDNF/PGA animals during the 7 day timeframe. Further investigation is required to determine whether significant behavioural improvement can be achieved at a longer timeframe.


Subject(s)
Amniotic Fluid/cytology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Stem Cell Transplantation , Stem Cells/physiology , Animals , Brain Injuries/pathology , Brain Injuries/physiopathology , Brain Injuries/therapy , Cell Survival , Cells, Cultured , Female , Gene Expression , Humans , Hydrogen Peroxide/pharmacology , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Motor Cortex/pathology , Neural Stem Cells/physiology , Oxidants/pharmacology , Oxidative Stress , Prostheses and Implants , Psychomotor Performance , Tissue Scaffolds
8.
Biochem Cell Biol ; 91(5): 271-86, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24032676

ABSTRACT

There is a need for improved therapy for acquired brain injury, which has proven resistant to treatment by numerous drugs in clinical trials and continues to represent one of the leading causes of disability worldwide. Research into cell-based therapies for the treatment of brain injury is growing rapidly, but the ideal cell source has yet to be determined. Subpopulations of cells found in amniotic fluid, which is readily obtained during routine amniocentesis, can be easily expanded in culture, have multipotent differentiation capacity, are non-tumourigenic, and avoid the ethical complications associated with embryonic stem cells, making them a promising cell source for therapeutic purposes. Beneficial effects of amniotic fluid cell transplantation have been reported in various models of nervous system injury. However, evidence that amniotic fluid cells can differentiate into mature, functional neurons in vivo and incorporate into the existing circuitry to replace lost or damaged neurons is lacking. The mechanisms by which amniotic fluid cells improve outcomes after experimental nervous system injury remain unclear. However, studies reporting the expression and release of neurotrophic, angiogenic, and immunomodulatory factors by amniotic fluid cells suggest they may provide neuroprotection and (or) stimulate endogenous repair and remodelling processes in the injured nervous system. In this paper, we address recent research related to the neuronal differentiation of amniotic fluid-derived cells, the therapeutic efficacy of these cells in animal models of nervous system injury, and the possible mechanisms mediating the positive outcomes achieved by amniotic fluid cell transplantation.


Subject(s)
Amniotic Fluid/cytology , Brain Injuries/therapy , Cell- and Tissue-Based Therapy/methods , Multipotent Stem Cells/transplantation , Amniocentesis , Animals , Cell Differentiation , Humans , Mice , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Neurons/cytology , Stroke/therapy , Tissue Engineering/methods
10.
Stem Cells Int ; 2012: 721538, 2012.
Article in English | MEDLINE | ID: mdl-23093978

ABSTRACT

The amniotic membrane (AM) and amniotic fluid (AF) have a long history of use in surgical and prenatal diagnostic applications, respectively. In addition, the discovery of cell populations in AM and AF which are widely accessible, nontumorigenic and capable of differentiating into a variety of cell types has stimulated a flurry of research aimed at characterizing the cells and evaluating their potential utility in regenerative medicine. While a major focus of research has been the use of amniotic membrane and fluid in tissue engineering and cell replacement, AM- and AF-derived cells may also have capabilities in protecting and stimulating the repair of injured tissues via paracrine actions, and acting as vectors for biodelivery of exogenous factors to treat injury and diseases. Much progress has been made since the discovery of AM and AF cells with stem cell characteristics nearly a decade ago, but there remain a number of problematic issues stemming from the inherent heterogeneity of these cells as well as inconsistencies in isolation and culturing methods which must be addressed to advance the field towards the development of cell-based therapies. Here, we provide an overview of the recent progress and future perspectives in the use of AM- and AF-derived cells for therapeutic applications.

11.
J Neurosci Res ; 90(12): 2362-77, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22987726

ABSTRACT

Nitric oxide (NO) plays a key role in neurogenesis as a regulator of cell proliferation and differentiation. NO is synthesized from the amino acid L-arginine by nitric oxide synthases (NOS1, NOS2, and NOS3), which are encoded by separate genes and display different tissue distributions. We used an in vitro model of RA-induced neural differentiation of NT2 cells to examine which of the three NO-synthesizing enzymes is involved in this process. The results revealed a transient induction of NOS3 (known as the constitutively expressed endothelial nitric oxide synthase; eNOS) during the time course of the RA treatment. The peak of gene expression and the nuclear presence of NOS3 protein coincided with cell cycle exit of NT2-derived neuronal precursors. The subsequent analysis of cytosine methylation and histone H3 acetylation of the human NOS3 5' regulatory sequences indicated that epigenetic modifications, especially upstream of the proximal promoter (-734 to -989, relative to exon 2 TSS at +1), were also taking place. NOS1 was expressed only in the differentiated neurons (NT2-N), whereas NOS2 was not expressed at all in this cellular model. Thus, a burst of NO production, possibly required to inhibit neural cell proliferation, was generated by the transient expression of NOS3. This pattern of gene expression, in turn, required epigenetic remodeling of its regulatory region.


Subject(s)
Nerve Tissue Proteins/physiology , Neurogenesis/drug effects , Nitric Oxide Synthase Type III/physiology , Tretinoin/pharmacology , 5' Untranslated Regions/genetics , Acetylation , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Line, Tumor/cytology , Cell Line, Tumor/drug effects , Cell Nucleus/enzymology , Chromatin Immunoprecipitation , CpG Islands/genetics , DNA Methylation , Enzyme Induction/drug effects , Gene Expression Regulation, Developmental/drug effects , Histones/metabolism , Humans , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurogenesis/physiology , Neuroglia/cytology , Neurons/cytology , Nitric Oxide/physiology , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type I/biosynthesis , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/genetics , Ornithine/analogs & derivatives , Ornithine/pharmacology , Protein Processing, Post-Translational , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Teratocarcinoma/pathology , Triazenes/pharmacology
12.
Stem Cells Int ; 2012: 607161, 2012.
Article in English | MEDLINE | ID: mdl-22792116

ABSTRACT

The usage of stem cells is a promising strategy for the repair of damaged tissue in the injured brain. Recently, amniotic fluid (AF) cells have received a lot of attention as an alternative source of stem cells for cell-based therapies. However, the success of this approach relies significantly on proper interactions between graft and host tissue. In particular, the reestablishment of functional brain networks requires formation of gap junctions, as a key step to provide sufficient intercellular communication. In this study, we show that AF cells express high levels of CX43 (GJA1) and are able to establish functional gap junctions with cortical cultures. Furthermore, we report an induction of Cx43 expression in astrocytes following injury to the mouse motor cortex and demonstrate for the first time CX43 expression at the interface between implanted AF cells and host brain cells. These findings suggest that CX43-mediated intercellular communication between AF cells and cortical astrocytes may contribute to the reconstruction of damaged tissue by mediating modulatory, homeostatic, and protective factors in the injured brain and hence warrants further investigation.

13.
J Neurosci Methods ; 205(1): 17-27, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22209770

ABSTRACT

Bone morphogenetic protein 7 (BMP7), a member of the transforming growth factor ß (TGF-ß) superfamily, plays important roles in the development of various tissues and organs in mouse and human. In particular, BMP7 is critical for the formation of the nervous system and it is considered to have therapeutic potential in brain injury and stroke. One approach to make BMP7 more suitable for therapeutic purposes is the development of efficient vectors that allow the consistent, reliable and cost-effective production of the BMP7 protein. In this study, we developed an efficient BMP7 delivery system, using a third generation lentiviral vector to produce functional BMP7 protein. The lentiviral transduction of several human cell types, including human embryonic kidney 293 (HEK293) cells, amniotic fluid cells, NTera2 neurons (NT2-N) and primary neuronal cultures resulted in BMP7 expression. The production of BMP7 protein was achieved for at least 4 weeks post-transduction, as determined by enzyme-linked immunosorbent assay (ELISA). SMAD phosphorylation and neuronal differentiation assays verified the bioactivity and functionality of the lentiviral-based BMP7 protein, respectively. In addition, the intracerebroventricular injection of the lentivirus resulted in exogenous BMP7 expression in both neurons and astrocytes in the mouse brain. Taken together, this gene delivery system provides a reliable source of functional BMP7 protein for future in vitro and in vivo studies.


Subject(s)
Bone Morphogenetic Protein 7/biosynthesis , Gene Transfer Techniques , Lentivirus/genetics , Transfection/methods , Amniotic Fluid/cytology , Animals , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cells, Cultured , DNA, Complementary/administration & dosage , DNA, Complementary/genetics , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Green Fluorescent Proteins , Humans , Immunohistochemistry , Injections, Intraventricular , Mice , Neurons/metabolism , Neurons/physiology , Pregnancy , Smad Proteins/biosynthesis , Smad Proteins/genetics , Transduction, Genetic
14.
Analyst ; 136(8): 1620-6, 2011 Apr 21.
Article in English | MEDLINE | ID: mdl-21369597

ABSTRACT

Brain injury can lead to irreversible tissue loss and functional deficit along with significant health care costs. Raman spectroscopy can be used as a non-invasive technique to provide detailed information on the molecular composition of diseased and damaged tissues. This technique was used to examine acute mouse brain injury, focusing on the motor cortex, a region directly involved in controlling execution of movement. The spectral profile obtained from the injured brain tissue revealed a markedly different signature, particularly in the amide I and amide III vibrational region when compared to that of healthy brain tissue. Most noticeably, there was a significant reduction of the amide I vibration at the acute injury site and the appearance of two distinct features at 1586 and 1618 cm(-1). Complementary immunohistochemical analysis of the injured brain tissue showed an abundant expression of Caspase 3 (a cysteine protease marker used for apoptosis), suggesting that the injury-induced specific Raman shifts may be correlated with cell death. Taken together, this study demonstrates that Raman spectroscopy can play an important role in detecting the changes that occur in the injured brain and provide a possible technology for monitoring the recovery process.


Subject(s)
Brain Injuries/pathology , Spectrum Analysis, Raman/methods , Amides/chemistry , Animals , Brain Injuries/enzymology , Caspase 3/metabolism , Discriminant Analysis , Mice , Principal Component Analysis
15.
Neural Dev ; 5: 31, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21122105

ABSTRACT

We previously identified four functionally distinct human NUMB isoforms. Here, we report the identification of two additional isoforms and propose a link between the expression of these isoforms and cancer. These novel isoforms, NUMB5 and NUMB6, lack exon 10 and are expressed in cells known for polarity and migratory behavior, such as human amniotic fluid cells, glioblastoma and metastatic tumor cells. RT-PCR and luciferase assays demonstrate that NUMB5 and NUMB6 are less antagonistic to NOTCH signaling than other NUMB isoforms. Immunocytochemistry analyses show that NUMB5 and NUMB6 interact and complex with CDC42, vimentin and the CDC42 regulator IQGAP1 (IQ (motif) GTPase activating protein 1). Furthermore, the ectopic expression of NUMB5 and NUMB6 induces the formation of lamellipodia (NUMB5) and filopodia (NUMB6) in a CDC42- and RAC1-dependent manner. These results are complemented by in vitro and in vivo studies, demonstrating that NUMB5 and NUMB6 alter the migratory behavior of cells. Together, these novel isoforms may play a role in further understanding the NUMB function in development and cancer.


Subject(s)
Fetal Development/physiology , Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Neoplasms/genetics , Nerve Tissue Proteins/genetics , Signal Transduction/physiology , Animals , Blotting, Western , Cell Movement/genetics , Cell Polarity/genetics , Chick Embryo , Gene Expression Profiling , Humans , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Neoplasms/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells , Neurogenesis/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Reverse Transcriptase Polymerase Chain Reaction
16.
Stem Cell Rev Rep ; 6(4): 677-84, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20574714

ABSTRACT

The NOTCH signaling pathway plays important roles in stem cell maintenance, cell-fate determination and differentiation during development. Following ligand binding, the cleaved NOTCH intracellular domain (NICD) interacts directly with the recombinant signal binding protein for immunoglobulin kappa J region (RBPJ) transcription factor and the resulting complex targets gene expression in the nucleus. To date, four human RBPJ isoforms have been described in Entrez Gene, varying in the first 5'coding exons. Using an improved protocol, we were able to further identify all four known and five novel RBPJ transcript variants in human amniotic fluid (AF) cells, a cell type known for its stem cell characteristics. In addition, we used human embryonal carcinoma (EC) NTera2/D1 (NT2) cells and NT2-derived neuron and astrocytes to compare the expression pattern of RBPJ transcripts. Further examination of RBPJ transcripts showed that the novel splice variants contain open reading frames in-frame with the known isoforms, suggesting that they can putatively generate similar function proteins. All known and novel RBPJ transcripts contain the putative nuclear localization signal (NLS), an important component of RBPJ-mediated gene regulation.


Subject(s)
Amniotic Fluid/cytology , Amniotic Fluid/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Cell Line, Tumor , Cells, Cultured , Humans , Immunohistochemistry , Protein Isoforms/genetics , Reverse Transcriptase Polymerase Chain Reaction
17.
Stem Cell Rev Rep ; 6(2): 199-214, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20221716

ABSTRACT

Recently, human amniotic fluid (AF) cells have attracted a great deal of attention as an alternative cell source for transplantation and tissue engineering. AF contains a variety of cell types derived from fetal tissues, of which a small percentage is believed to represent stem cell sub-population(s). In contrast to human embryonic stem (ES) cells, AF cells are not subject to extensive legal or ethical considerations; nor are they limited by lineage commitment characteristic of adult stem cells. However, to become therapeutically valuable, better protocols for the isolation of AF stem cell sub-populations need to be developed. This study was designed to examine the molecular components involved in self-renewal, neural commitment and differentiation of AF cells obtained at different gestational ages. Our results showed that, although morphologically heterogeneous, AF cells derived from early gestational periods ubiquitously expressed KERATIN 8 (K8), suggesting that the majority of these cells may have an epithelial origin. In addition, AF cells expressed various components of NOTCH signaling (ligands, receptors and target genes), a pathway involved in stem cell maintenance, determination and differentiation. A sub-population of K8 positive cells (<10%) co-expressed NESTIN, a marker detected in the neuroepithelium, neural stem cells and neural progenitors. Throughout the gestational periods, a much smaller AF cell sub-population (<1%) expressed pluripotency markers, OCT4a, NANOG and SOX2, from which SOX2 positive AF cells could be isolated through single cell cloning. The SOX2 expressing AF clones showed the capacity to give rise to a neuron-like phenotype in culture, expressing neuronal markers such as MAP2, NFL and NSE. Taken together, our findings demonstrated the presence of fetal cells with stem cell characteristics in the amniotic fluid, highlighting the need for further research on their biology and clinical applications.


Subject(s)
Amniotic Fluid/cytology , Stem Cells/cytology , Stem Cells/metabolism , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Female , Flow Cytometry , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Keratin-8/genetics , Keratin-8/metabolism , Models, Biological , Nanog Homeobox Protein , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Signal Transduction
18.
J Neurosci Methods ; 186(1): 60-7, 2010 Jan 30.
Article in English | MEDLINE | ID: mdl-19903493

ABSTRACT

Neuro 2A (N2a) is a mouse neural crest-derived cell line that has been extensively used to study neuronal differentiation, axonal growth and signaling pathways. A convenient characteristic of these cells is their ability to differentiate into neurons within a few days. However, most differentiation methods reported for N2a cells do not provide information about the neuronal types obtained after each treatment. In this study, we evaluated the generation of N2a dopamine neurons following treatment with a number of factors known to induce neuronal differentiation. Our results showed that N2a cells express Nurr-related factor 1 (Nurr1) and produce low levels of tyrosine hydroxylase (TH) and dopamine. Both TH and dopamine levels were significantly enhanced in the presence of dibutyryl cyclic adenosine monophosphate (dbcAMP), as evidenced by Western blot, immunocytochemistry and high performance liquid chromatography (HPLC). In contrast to dbcAMP, other factors such as transforming growth factor beta1 (TGF beta 1), bone morphogenetic protein 4 (BMP4), glial cell-derived neurotrophic factor (GDNF) and retinoic acid (RA) did not increase TH expression. Further investigation confirmed that the effect of dbcAMP on production of TH-positive neurons was mediated through cyclic AMP (cAMP) responsive element binding protein (CREB) and it was antagonized by RA. Thus, although various treatments can be used to generate N2a neurons, only dbcAMP significantly enhanced the formation of dopamine neurons. Taken together, this study provided a simple and reliable method to generate dopamine neurons for rapid and efficient physiological and pharmacological assays.


Subject(s)
Cell Differentiation/physiology , Dopamine/metabolism , Neurogenesis/physiology , Neurons/metabolism , Stem Cells/metabolism , Animals , Bucladesine/pharmacology , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Mice , Nerve Growth Factors/pharmacology , Neurogenesis/drug effects , Neurons/cytology , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Tretinoin/metabolism , Tretinoin/pharmacology , Tyrosine 3-Monooxygenase/metabolism
19.
Exp Cell Res ; 316(1): 68-77, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19744480

ABSTRACT

Myosin phosphatase target subunit 1 (MYPT1), together with catalytic subunit of type1 delta isoform (PP1cdelta) and a small 20-kDa regulatory unit (M20), form a heterotrimeric holoenzyme, myosin phosphatase (MP), which is responsible for regulating the extent of myosin light chain phosphorylation. Here we report the identification and characterization of a molecular interaction between Seven in absentia homolog 2 (SIAH2) and MYPT1 that resulted in the proteasomal degradation of the latter in mammalian cells, including neurons and glia. The interaction involved the substrate binding domain of SIAH2 (aa 116-324) and a central region of MYPT1 (aa 445-632) containing a degenerate consensus Siah-binding motif RLAYVAP (aa 493-499) evolutionally conserved from fish to humans. These findings suggest a novel mechanism whereby the ability of MP to modulate myosin light chain might be regulated by the degradation of its targeting subunit MYPT1 through the SIAH2-ubiquitin-proteasomal pathway. In this manner, the turnover of MYPT1 would serve to limit the duration and/or magnitude of MP activity required to achieve a desired physiological effect.


Subject(s)
Myosin-Light-Chain Phosphatase/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Animals , Astrocytes/metabolism , Binding Sites/physiology , Cell Line , Cell Line, Tumor , Cells, Cultured , Consensus Sequence/physiology , Cysteine Proteinase Inhibitors/pharmacology , Cytoplasm/metabolism , Gene Expression/genetics , Humans , Mice , Mice, Inbred Strains , Molecular Sequence Data , Mutation/physiology , Myosin-Light-Chain Phosphatase/genetics , Neurons/metabolism , Nuclear Proteins/genetics , Peptide Fragments/genetics , Peptide Fragments/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Protein Binding/physiology , Protein Interaction Domains and Motifs/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Transfection , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases/genetics
20.
Biochem Biophys Res Commun ; 382(1): 85-90, 2009 Apr 24.
Article in English | MEDLINE | ID: mdl-19265675

ABSTRACT

Adrenergic receptors are a key component of nanoscale multiprotein complexes that are responsible for controlling the beat rate in a mammalian heart. We demonstrate the ability of near-field scanning optical microscopy (NSOM) to visualize beta(2)-adrenergic receptors (beta(2)AR) fused to the GFP analogue Venus at the nanoscale on HEK293 cells. The expression of the beta(2)AR-Venus fusion protein was tightly controlled using a tetracycline-induced promoter. Both the size and density of the observed nanoscale domains are dependent on the level of induction and thus the level of protein expression. At concentrations between 100 and 700 ng/ml of inducer doxycycline, the size of domains containing the beta(2)AR-Venus fusion protein appears to remain roughly constant, but the number of domains per cell increase. At 700 ng/ml doxycycline the functional receptors are organized into domains with an average diameter of 150 nm with a density similar to that observed for the native protein on primary murine cells. By contrast, larger micron-sized domains of beta(2)AR are observed in the membrane of the HEK293 cells that stably overexpress beta(2)AR-GFP and beta(2)AR-eYFP. We conclude that precise chemical control of gene expression is highly advantageous for the use beta(2)AR-Venus fusion proteins as models for beta(2)AR function. These observations are critical for designing future cell models and assays based on beta(2)AR, since the receptor biology is consistent with a relatively low density of nanoscale receptor domains.


Subject(s)
Bacterial Proteins/metabolism , Luminescent Proteins/metabolism , Receptors, Adrenergic, beta-2/metabolism , Recombinant Fusion Proteins/metabolism , Bacterial Proteins/biosynthesis , Bacterial Proteins/ultrastructure , Cell Line , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Humans , Luminescent Proteins/biosynthesis , Luminescent Proteins/ultrastructure , Microscopy, Confocal/methods , Protein Structure, Tertiary , Receptors, Adrenergic, beta-2/biosynthesis , Receptors, Adrenergic, beta-2/ultrastructure , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...