Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Proteome Res ; 16(2): 481-493, 2017 02 03.
Article in English | MEDLINE | ID: mdl-28152600

ABSTRACT

We undertook an unbiased metabolite profiling of fibroblasts from schizophrenia patients and healthy controls to identify metabolites and pathways that are dysregulated in disease, seeking to gain new insights into the disease biology of schizophrenia and to discover potential disease-related biomarkers. We measured polar and nonpolar metabolites in the fibroblasts under normal conditions and under two stressful physiological perturbations: growth in low-glucose media and exposure to the steroid hormone dexamethasone. We found that metabolites that were significantly different between schizophrenia and control subjects showed separation of the two groups by partial least-squares discriminant analysis methods. This separation between schizophrenia and healthy controls was more robust with metabolites identified under the perturbation conditions. The most significant individual metabolite differences were also found in the perturbation experiments. Metabolites that were significantly different between schizophrenia and healthy controls included a number of plasmalogens and phosphatidylcholines. We present these results in the context of previous reports of metabolic profiling of brain tissue and plasma in schizophrenia. These results show the applicability of metabolite profiling under stressful perturbations to reveal cellular pathways that may be involved in disease biology.


Subject(s)
Fibroblasts/metabolism , Metabolome , Phosphatidylcholines/metabolism , Plasmalogens/metabolism , Schizophrenia/metabolism , Stress, Physiological , Adult , Antipsychotic Agents/therapeutic use , Biomarkers/metabolism , Case-Control Studies , Culture Media/pharmacology , Dexamethasone/pharmacology , Discriminant Analysis , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Glucocorticoids/pharmacology , Glucose/deficiency , Glucose/pharmacology , Humans , Least-Squares Analysis , Male , Middle Aged , Primary Cell Culture , Schizophrenia/drug therapy , Schizophrenia/pathology
2.
Mol Neuropsychiatry ; 2(2): 97-106, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27606323

ABSTRACT

Many studies suggest the presence of aberrations in cellular metabolism in bipolar disorder. We studied the metabolome in bipolar disorder to gain insight into cellular pathways that may be dysregulated in bipolar disorder and to discover evidence of novel biomarkers. We measured polar and nonpolar metabolites in fibroblasts from subjects with bipolar I disorder and matched healthy control subjects, under normal conditions and with two physiologic perturbations: low-glucose media and exposure to the stress-mediating hormone dexamethasone. Metabolites that were significantly different between bipolar and control subjects showed distinct separation by principal components analysis methods. The most statistically significant findings were observed in the perturbation experiments. The metabolite with the lowest p value in both the low-glucose and dexamethasone experiments was α-aminoadipate, whose intracellular level was consistently lower in bipolar subjects. Our study implicates α-aminoadipate as a possible biomarker in bipolar disorder that manifests under cellular stress. This is an intriguing finding given the known role of α-aminoadipate in the modulation of kynurenic acid in the brain, especially as abnormal kynurenic acid levels have been implicated in bipolar disorder.

3.
Mol Cell Neurosci ; 73: 96-103, 2016 06.
Article in English | MEDLINE | ID: mdl-26777134

ABSTRACT

Schizophrenia and bipolar disorder are complex psychiatric disorders that present unique challenges in the study of disease biology. There are no objective biological phenotypes for these disorders, which are characterized by complex genetics and prominent roles for gene-environment interactions. The study of the neurobiology underlying these severe psychiatric disorders has been hindered by the lack of access to the tissue of interest - neurons from patients. The advent of reprogramming methods that enable generation of induced pluripotent stem cells (iPSCs) from patient fibroblasts and peripheral blood mononuclear cells has opened possibilities for new approaches to study relevant disease biology using iPSC-derived neurons. While early studies with patient iPSCs have led to promising and intriguing leads, significant hurdles remain in our attempts to capture the complexity of these disorders in vitro. We present here an overview of studies to date of schizophrenia and bipolar disorder using iPSC-derived neuronal cells and discuss potential future directions that can result in the identification of robust and valid cellular phenotypes that in turn can lay the groundwork for meaningful clinical advances.


Subject(s)
Bipolar Disorder/pathology , Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/cytology , Schizophrenia/pathology , Bipolar Disorder/genetics , Bipolar Disorder/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Phenotype , Precision Medicine/methods , Schizophrenia/genetics , Schizophrenia/metabolism
4.
ACS Chem Biol ; 10(3): 883-90, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25546293

ABSTRACT

We examined the effects of isoform-specific histone deacetylase (HDAC) inhibitors on ß-catenin posttranslational modifications in neural progenitor cells (NPCs) derived from human induced pluripotent stem cells (iPSCs). ß-catenin is a multifunctional protein with important roles in the developing and adult central nervous system. Activation of the Wnt pathway results in stabilization and nuclear translocation of ß-catenin, resulting in activation of multiple target genes. In addition, ß-catenin forms a complex with cadherins at the plasma membrane as part of the adherens junctions. The N-terminus of ß-catenin has phosphorylation, ubiquitination, and acetylation sites that regulate its stability and signaling. In the absence of a Wnt signal, Ser33, Ser37, and Thr41 are constitutively phosphorylated by glycogen synthase kinase 3ß (GSK3ß). ß-Catenin phosphorylated at these sites is recognized by ß-transducin repeat-containing protein (ßTrCP), which results in ubiquitination and degradation by the ubiquitin-proteasome pathway. The N-terminal regulatory domain of ß-catenin also includes Ser45, a phosphorylation site for Casein Kinase 1α (CK1α) and Lys49, which is acetylated by the acetyltransferase p300/CBP-associated factor (PCAF). The relevance of Lys49 acetylation and Ser45 phosphorylation to the function of ß-catenin is an active area of investigation. We find that HDAC6 inhibitors increase Lys49 acetylation and Ser45 phosphorylation but do not affect Ser33, Ser37, and Thr41 phosphorylation. Lys49 acetylation results in decreased ubiquitination of ß-catenin in the presence of proteasome inhibition. While increased Lys49 acetylation does not affect total levels of ß-catenin, it results in increased membrane localization of ß-catenin.


Subject(s)
Cell Membrane/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Neurons/drug effects , Protein Processing, Post-Translational , beta Catenin/metabolism , Acetylation , Binding Sites , Cell Differentiation , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cells, Cultured , Gene Expression , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Lysine/chemistry , Lysine/metabolism , Models, Molecular , Neurons/metabolism , Neurons/ultrastructure , Phosphorylation , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Signal Transduction , Structure-Activity Relationship , Ubiquitination , beta Catenin/chemistry , beta Catenin/genetics
5.
Tissue Eng Part A ; 20(15-16): 2200-12, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24498910

ABSTRACT

Drug-induced liver injury is a major cause of drug development failures and postmarket withdrawals. In vitro models that incorporate primary hepatocytes have been shown to be more predictive than model systems which rely on liver microsomes or hepatocellular carcinoma cell lines. Methods to phenotypically stabilize primary hepatocytes ex vivo often rely on mimicry of hepatic microenvironmental cues such as cell-cell interactions and cell-matrix interactions. In this work, we sought to incorporate phenotypically stable hepatocytes into three-dimensional (3D) microtissues, which, in turn, could be deployed in drug-screening platforms such as multiwell plates and diverse organ-on-a-chip devices. We first utilize micropatterning on collagen I to specify cell-cell interactions in two-dimensions, followed by collagenase digestion to produce well-controlled aggregates for 3D encapsulation in polyethylene glycol (PEG) diacrylate. Using this approach, we examined the influence of homotypic hepatocyte interactions and composition of the encapsulating hydrogel, and achieved the maintenance of liver-specific function for over 50 days. Optimally preaggregated structures were subsequently encapsulated using a microfluidic droplet-generator to produce 3D microtissues. Interactions of engineered hepatic microtissues with drugs was characterized by flow cytometry, and yielded both induction of P450 enzymes in response to prototypic small molecules and drug-drug interactions that give rise to hepatotoxicity. Collectively, this study establishes a pipeline for the manufacturing of 3D hepatic microtissues that exhibit stabilized liver-specific functions and can be incorporated into a wide array of emerging drug development platforms.


Subject(s)
Cell Communication/drug effects , Hepatocytes/cytology , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Tissue Engineering/methods , 3T3 Cells , Acetaminophen/toxicity , Albumins/metabolism , Animals , Cells, Cultured , Cells, Immobilized/cytology , Cells, Immobilized/drug effects , Cells, Immobilized/metabolism , Collagen/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Mice , Microfluidics , Rats, Inbred Lew
6.
Lab Chip ; 13(10): 1969-78, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23563587

ABSTRACT

The cancer microenvironment, which incorporates interactions with stromal cells, extracellular matrix (ECM), and other tumor cells in a 3-dimensional (3D) context, has been implicated in every stage of cancer development, including growth of the primary tumor, metastatic spread, and response to treatment. Our understanding of the tumor microenvironment and our ability to develop new therapies would greatly benefit from tools that allow us to systematically probe microenvironmental cues within a 3D context. Here, we leveraged recent advances in microfluidic technology to develop a platform for high-throughput fabrication of tunable cellular microniches ("microtissues") that allow us to probe tumor cell response to a range of microenvironmental cues, including ECM, soluble factors, and stromal cells, all in 3D. We further combine this tunable microniche platform with rapid, flow-based population level analysis (n > 500), which permits analysis and sorting of microtissue populations both pre- and post-culture by a range of parameters, including proliferation and homotypic or heterotypic cell density. We used this platform to demonstrate differential responses of lung adenocarcinoma cells to a selection of ECM molecules and soluble factors. The cells exhibited enhanced or reduced proliferation when encapsulated in fibronectin- or collagen-1-containing microtissues, respectively, and they showed reduced proliferation in the presence of TGF-ß, an effect that we did not observe in monolayer culture. We also measured tumor cell response to a panel of drug targets and found, in contrast to monolayer culture, specific sensitivity of tumor cells to TGFßR2 inhibitors, implying that TGF-ß has an anti-proliferative affect that is unique to the 3D context and that this effect is mediated by TGFßR2. These findings highlight the importance of the microenvironmental context in therapeutic development and that the platform we present here allows the high-throughput study of tumor response to drugs as well as basic tumor biology in well-defined microenvironmental niches.


Subject(s)
Cell Culture Techniques/methods , Tumor Microenvironment , Animals , Cell Culture Techniques/instrumentation , Cell Line, Tumor , Collagen/pharmacology , Extracellular Matrix/metabolism , Fibronectins/pharmacology , Humans , Mice , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Rats , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Stromal Cells/cytology , Stromal Cells/drug effects , Transforming Growth Factor beta/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...