Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nature ; 567(7749): 535-539, 2019 03.
Article in English | MEDLINE | ID: mdl-30867594

ABSTRACT

Chemical modifications of histones can mediate diverse DNA-templated processes, including gene transcription1-3. Here we provide evidence for a class of histone post-translational modification, serotonylation of glutamine, which occurs at position 5 (Q5ser) on histone H3 in organisms that produce serotonin (also known as 5-hydroxytryptamine (5-HT)). We demonstrate that tissue transglutaminase 2 can serotonylate histone H3 tri-methylated lysine 4 (H3K4me3)-marked nucleosomes, resulting in the presence of combinatorial H3K4me3Q5ser in vivo. H3K4me3Q5ser displays a ubiquitous pattern of tissue expression in mammals, with enrichment observed in brain and gut, two organ systems responsible for the bulk of 5-HT production. Genome-wide analyses of human serotonergic neurons, developing mouse brain and cultured serotonergic cells indicate that H3K4me3Q5ser nucleosomes are enriched in euchromatin, are sensitive to cellular differentiation and correlate with permissive gene expression, phenomena that are linked to the potentiation of TFIID4-6 interactions with H3K4me3. Cells that ectopically express a H3 mutant that cannot be serotonylated display significantly altered expression of H3K4me3Q5ser-target loci, which leads to deficits in differentiation. Taken together, these data identify a direct role for 5-HT, independent from its contributions to neurotransmission and cellular signalling, in the mediation of permissive gene expression.


Subject(s)
Gene Expression Regulation , Histones/chemistry , Histones/metabolism , Lysine/metabolism , Protein Processing, Post-Translational , Serotonin/metabolism , Transcription Factor TFIID/metabolism , Animals , Cell Differentiation , Cell Line , Female , GTP-Binding Proteins/metabolism , Glutamine/chemistry , Glutamine/metabolism , Humans , Methylation , Mice , Mice, Inbred C57BL , Protein Binding , Protein Glutamine gamma Glutamyltransferase 2 , Serotonergic Neurons/cytology , Transglutaminases/metabolism
2.
Cell ; 156(5): 1114-1114.e1, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24581504
3.
Mol Psychiatry ; 26(7): 2753-2763, 2021 07.
Article in English | MEDLINE | ID: mdl-33767349

ABSTRACT

The serotonergic system in the human brain modulates several physiological processes, and altered serotonergic neurotransmission has been implicated in the neuropathology of several psychiatric disorders. The study of serotonergic neurotransmission in psychiatry has long been restricted to animal models, but advances in cell reprogramming technology have enabled the generation of serotonergic neurons from patient-induced pluripotent stem cells (iPSCs). While iPSC-derived human serotonergic neurons offer the possibility to study serotonin (5-HT) release and uptake, particularly by 5-HT-modulating drugs such as selective serotonin reuptake inhibitors (SSRIs), a major limitation is the inability to reliably quantify 5-HT secreted from neurons in vitro. Herein, we address this technical gap via a novel sensing technology that couples 5-HT-specific DNA aptamers into nanopores (glass nanopipettes) with orifices of ~10 nm to detect 5-HT in complex neuronal culture medium with higher selectivity, sensitivity, and stability than existing methods. The 5-HT aptamers undergo conformational rearrangement upon target capture and serve as gatekeepers of ionic flux through the nanopipette opening. We generated human serotonergic neurons in vitro and detected secreted 5-HT using aptamer-coated nanopipettes in a low nanomolar range, with the possibility of detecting significantly lower (picomolar) concentrations. Furthermore, as a proof of concept, we treated human serotonergic neurons in vitro with the SSRI citalopram and detected a significant increase in extracellular 5-HT using the aptamer-modified nanopipettes. We demonstrate the utility of such methods for 5-HT detection, raising the possibility of fast quantification of neurotransmitters secreted from patient-derived live neuronal cells.


Subject(s)
Serotonergic Neurons , Serotonin , Animals , Brain , Citalopram/pharmacology , Humans , Selective Serotonin Reuptake Inhibitors/pharmacology
4.
Mol Psychiatry ; 24(6): 808-818, 2019 06.
Article in English | MEDLINE | ID: mdl-30903001

ABSTRACT

Disrupted serotonergic neurotransmission has long been implicated in major depressive disorder (MDD), for which selective serotonin reuptake inhibitors (SSRIs) are the first line of treatment. However, a significant percentage of patients remain SSRI-resistant and it is unclear whether and how alterations in serotonergic neurons contribute to SSRI resistance in these patients. Induced pluripotent stem cells (iPSCs) facilitate the study of patient-specific neural subtypes that are typically inaccessible in living patients, enabling the discovery of disease-related phenotypes. In our study of a well-characterized cohort of over 800 MDD patients, we generated iPSCs and serotonergic neurons from three extreme SSRI-remitters (R) and SSRI-nonremitters (NR). We studied serotonin (5-HT) biochemistry and observed no significant differences in 5-HT release and reuptake or in genes related to 5-HT biochemistry. NR patient-derived serotonergic neurons exhibited altered neurite growth and morphology downstream of lowered expression of key Protocadherin alpha genes as compared to healthy controls and Rs. Furthermore, knockdown of Protocadherin alpha genes directly regulated iPSC-derived neurite length and morphology. Our results suggest that intrinsic differences in serotonergic neuron morphology and the resulting circuitry may contribute to SSRI resistance in MDD patients.


Subject(s)
Depressive Disorder, Treatment-Resistant/drug therapy , Depressive Disorder, Treatment-Resistant/physiopathology , Serotonin/metabolism , Adult , Antidepressive Agents/therapeutic use , Cohort Studies , Depressive Disorder, Major/drug therapy , Female , Humans , Induced Pluripotent Stem Cells/drug effects , Middle Aged , Neurons , Serotonergic Neurons/physiology , Selective Serotonin Reuptake Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/therapeutic use , Synaptic Transmission
5.
Mol Psychiatry ; 24(6): 795-807, 2019 06.
Article in English | MEDLINE | ID: mdl-30700803

ABSTRACT

Selective serotonin reuptake inhibitors (SSRIs) are the most prescribed antidepressants. They regulate serotonergic neurotransmission, but it remains unclear how altered serotonergic neurotransmission may contribute to the SSRI resistance observed in approximately 30% of major depressive disorder (MDD) patients. Patient stratification based on pharmacological responsiveness and the use of patient-derived neurons may make possible the discovery of disease-relevant neural phenotypes. In our study from a large cohort of well-characterized MDD patients, we have generated induced pluripotent stem cells (iPSCs) from SSRI-remitters and SSRI-nonremitters. We studied serotonergic neurotransmission in patient forebrain neurons in vitro and observed that nonremitter patient-derived neurons displayed serotonin-induced hyperactivity downstream of upregulated excitatory serotonergic receptors, in contrast to what is seen in healthy and remitter patient-derived neurons. Our data suggest that postsynaptic forebrain hyperactivity downstream of SSRI treatment may play a role in SSRI resistance in MDD.


Subject(s)
Depressive Disorder, Treatment-Resistant/drug therapy , Depressive Disorder, Treatment-Resistant/physiopathology , Serotonin/metabolism , Adult , Akathisia, Drug-Induced/physiopathology , Antidepressive Agents/therapeutic use , Cohort Studies , Depressive Disorder, Major/drug therapy , Female , Humans , Induced Pluripotent Stem Cells/drug effects , Middle Aged , Neurons , Psychomotor Agitation/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/therapeutic use , Synaptic Transmission
6.
Cell Tissue Res ; 371(1): 161-170, 2018 01.
Article in English | MEDLINE | ID: mdl-28812143

ABSTRACT

Several lines of evidence implicate serotonin in the etiology of multiple psychiatric disorders, especially mood disorders, such as major depressive disorder (MDD) and bipolar disorder (BD). Much of our current understanding of biological mechanisms underlying serotonergic alterations in mood disorders comes from animal studies. Innovation in induced pluripotent stem cell and transdifferentiation technologies for deriving neurons from adult humans has enabled the study of disease-relevant cellular phenotypes in vitro. In this context, human serotonergic neurons can now be generated using three recently published methodologies. In this mini-review, we broadly discuss evidence linking altered serotonergic neurotransmission in MDD and BD and focus on recently published methods for generating human serotonergic neurons in vitro.


Subject(s)
Bipolar Disorder/physiopathology , Depressive Disorder, Major/physiopathology , Induced Pluripotent Stem Cells/physiology , Models, Neurological , Serotonergic Neurons/physiology , Serotonin/physiology , Adult , Animals , Cell Culture Techniques , Cell Transdifferentiation , Humans , Mice , Synaptic Transmission
7.
Bioessays ; 38(11): 1123-1129, 2016 11.
Article in English | MEDLINE | ID: mdl-27716980

ABSTRACT

Technologies for deriving human neurons in vitro have transformed our ability to study cellular and molecular components of human neurotransmission. Three groups, including our own, have recently published methods for efficiently generating human serotonergic neurons in vitro. Remarkably, serotonergic neurons derived from each method robustly produce serotonin, express raphe genes, are electrically active, and respond to selective serotonin reuptake inhibitors in vitro. Two of the methods utilize transdifferentiation technology by overexpressing key serotonergic transcription factors. The third and most recent method involves differentiating induced pluripotent stem cells (iPSCs) to serotonergic neurons using developmental patterning cues. In this mini-review, we briefly describe the developmental programs governing serotonergic specification in vivo and how they have been harnessed to achieve serotonergic differentiation in vitro. We discuss the distinct and overlapping features of the recently published methodologies and their value in the context of in vitro disease modeling. Also see the video abstract here.


Subject(s)
Cellular Reprogramming Techniques/methods , Serotonergic Neurons , Animals , Cell Differentiation , Cell Transdifferentiation , Humans , Induced Pluripotent Stem Cells/physiology , Intercellular Signaling Peptides and Proteins , Mice , Transcription Factors
8.
J Neurosci ; 33(3): 1179-89, 2013 Jan 16.
Article in English | MEDLINE | ID: mdl-23325254

ABSTRACT

The molecular mechanisms underlying the generation, maturation, and integration of new granule cells generated throughout life in the mammalian hippocampus remain poorly understood. Small Rho GTPases, such as Cdc42 and Rac1, have been implicated previously in neural stem/progenitor cell (NSPC) proliferation and neuronal maturation during embryonic development. Here we used conditional genetic deletion and virus-based loss-of-function approaches to identify temporally distinct functions for Cdc42 and Rac1 in adult hippocampal neurogenesis. We found that Cdc42 is involved in mouse NSPC proliferation, initial dendritic development, and dendritic spine maturation. In contrast, Rac1 is dispensable for early steps of neuronal development but is important for late steps of dendritic growth and spine maturation. These results establish cell-autonomous and stage-specific functions for the small Rho GTPases Cdc42 and Rac1 in the course of adult hippocampal neurogenesis.


Subject(s)
Hippocampus/metabolism , Neurogenesis/physiology , Neurons/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Cell Lineage/physiology , Cell Movement/physiology , Cell Proliferation , Dendrites/metabolism , Dendritic Spines/metabolism , Female , Mice , Mice, Transgenic , Rats , Rats, Inbred F344 , cdc42 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/genetics
9.
Proc Natl Acad Sci U S A ; 108(14): 5807-12, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21436036

ABSTRACT

Neural stem cells (NSCs) generate new granule cells throughout life in the mammalian hippocampus. Canonical Wnt signaling regulates the differentiation of NSCs towards the neuronal lineage. Here we identified the prospero-related homeodomain transcription factor Prox1 as a target of ß-catenin-TCF/LEF signaling in vitro and in vivo. Prox1 overexpression enhanced neuronal differentiation whereas shRNA-mediated knockdown of Prox1 impaired the generation of neurons in vitro and within the hippocampal niche. In contrast, Prox1 was not required for survival of adult-generated granule cells after they had matured, suggesting a role for Prox1 in initial granule cell differentiation but not in the maintenance of mature granule cells. The data presented here characterize a molecular pathway from Wnt signaling to a transcriptional target leading to granule cell differentiation within the adult brain and identify a stage-specific function for Prox1 in the process of adult neurogenesis.


Subject(s)
Cell Differentiation/physiology , Hippocampus/growth & development , Homeodomain Proteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Signal Transduction/physiology , Tumor Suppressor Proteins/metabolism , Wnt Proteins/metabolism , Animals , Base Sequence , Blotting, Western , Cells, Cultured , Chromatin Immunoprecipitation , DNA Primers/genetics , Hippocampus/cytology , Homeodomain Proteins/genetics , Immunohistochemistry , In Situ Hybridization , Luciferases , Mice , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Proteins/genetics
10.
IBRO Neurosci Rep ; 13: 420-425, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36386600

ABSTRACT

Norepinephrine (NE), and specific adrenoceptors, have been reported to influence distinct aspects of adult hippocampal neurogenesis, including latent stem cell activation, progenitor proliferation, and differentiation. These findings are predominantly based on the use of pharmacological approaches in both in vitro and in vivo systems. Here, we sought to assess the consequences of genetic ablation of NE on adult hippocampal neurogenesis, by examining dopamine ß hydroxylase knockout (Dbh -/-) mice, which lack NE from birth. We find that Dbh -/- mice exhibit no difference in adult hippocampal progenitor proliferation and survival. Further, the number of immature newborn neurons, labeled using stage-specific developmental markers within the hippocampal neurogenic niche, was also unaltered in Dbh -/- mice. In contrast, the noradrenergic neurotoxin DSP-4, which had previously been shown to reduce adult hippocampal neurogenesis in rats, also resulted in a decline in hippocampal progenitor proliferation in C57/Bl6N mice. These findings indicate that pharmacological lesioning of noradrenergic afferents in adulthood, but not the complete genetic loss of NE from birth, impairs adult hippocampal neurogenesis in mice.

11.
J Neurosci ; 30(3): 1096-109, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-20089918

ABSTRACT

Slow-onset adaptive changes that arise from sustained antidepressant treatment, such as enhanced adult hippocampal neurogenesis and increased trophic factor expression, play a key role in the behavioral effects of antidepressants. alpha(2)-Adrenoceptors contribute to the modulation of mood and are potential targets for the development of faster acting antidepressants. We investigated the influence of alpha(2)-adrenoceptors on adult hippocampal neurogenesis. Our results indicate that alpha(2)-adrenoceptor agonists, clonidine and guanabenz, decrease adult hippocampal neurogenesis through a selective effect on the proliferation, but not the survival or differentiation, of progenitors. These effects persist in dopamine beta-hydroxylase knock-out (Dbh(-/-)) mice lacking norepinephrine, supporting a role for alpha(2)-heteroceptors on progenitor cells, rather than alpha(2)-autoreceptors on noradrenergic neurons that inhibit norepinephrine release. Adult hippocampal progenitors in vitro express all the alpha(2)-adrenoceptor subtypes, and decreased neurosphere frequency and BrdU incorporation indicate direct effects of alpha(2)-adrenoceptor stimulation on progenitors. Furthermore, coadministration of the alpha(2)-adrenoceptor antagonist yohimbine with the antidepressant imipramine significantly accelerates effects on hippocampal progenitor proliferation, the morphological maturation of newborn neurons, and the increase in expression of brain derived neurotrophic factor and vascular endothelial growth factor implicated in the neurogenic and behavioral effects of antidepressants. Finally, short-duration (7 d) yohimbine and imipramine treatment results in robust behavioral responses in the novelty suppressed feeding test, which normally requires 3 weeks of treatment with classical antidepressants. Our results demonstrate that alpha(2)-adrenoceptors, expressed by progenitor cells, decrease adult hippocampal neurogenesis, while their blockade speeds up antidepressant action, highlighting their importance as targets for faster acting antidepressants.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists , Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Hippocampus/drug effects , Imipramine/pharmacology , Neurogenesis/drug effects , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Adrenergic beta-Agonists/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Antiparkinson Agents/pharmacology , Ascorbic Acid/pharmacology , Brain-Derived Neurotrophic Factor , Bromodeoxyuridine/metabolism , Cells, Cultured , Dopamine beta-Hydroxylase/genetics , Doublecortin Domain Proteins , Droxidopa/pharmacology , Drug Administration Schedule , Drug Interactions , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , In Vitro Techniques , Isoproterenol/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuropeptides/metabolism , Phenylephrine/pharmacology , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Wistar , Reaction Time/drug effects , Receptors, Adrenergic, alpha-2/physiology , Stem Cells/drug effects , Yohimbine/pharmacology
12.
Transl Psychiatry ; 11(1): 608, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34848679

ABSTRACT

Major depressive disorder (MDD) is a prevalent psychiatric disorder, and exposure to stress is a robust risk factor for MDD. Clinical data and rodent models have indicated the negative impact of chronic exposure to stress-induced hormones like cortisol on brain volume, memory, and cell metabolism. However, the cellular and transcriptomic changes that occur in the brain after prolonged exposure to cortisol are less understood. Furthermore, the astrocyte-specific contribution to cortisol-induced neuropathology remains understudied. Here, we have developed an in vitro model of "chronic stress" using human induced pluripotent stem cell (iPSC)-derived astrocytes treated with cortisol for 7 days. Whole transcriptome sequencing reveals differentially expressed genes (DEGs) uniquely regulated in chronic cortisol compared to acute cortisol treatment. Utilizing this paradigm, we examined the stress response transcriptome of astrocytes generated from MDD patient iPSCs. The MDD-specific DEGs are related to GPCR ligand binding, synaptic signaling, and ion homeostasis. Together, these data highlight the unique role astrocytes play in the central nervous system and present interesting genes for future study into the relationship between chronic stress and MDD.


Subject(s)
Depressive Disorder, Major , Induced Pluripotent Stem Cells , Astrocytes , Humans , Hydrocortisone , Ligands , Receptors, G-Protein-Coupled
13.
Stem Cell Reports ; 16(4): 825-835, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33667413

ABSTRACT

Bipolar disorder (BD) is characterized by cyclical mood shifts. Studies indicate that BD patients have a peripheral pro-inflammatory state and alterations in glial populations in the brain. We utilized an in vitro model to study inflammation-related phenotypes of astrocytes derived from induced pluripotent stem cells (iPSCs) generated from BD patients and healthy controls. BD astrocytes showed changes in transcriptome and induced a reduction in neuronal activity when co-cultured with neurons. IL-1ß-stimulated BD astrocytes displayed a unique inflammatory gene expression signature and increased secretion of IL-6. Conditioned medium from stimulated BD astrocytes reduced neuronal activity, and this effect was partially blocked by IL-6 inactivating antibody. Our results suggest that BD astrocytes are functionally less supportive of neuronal excitability and this effect is partially mediated by IL-6. We confirmed higher IL-6 in blood in a distinct cohort of BD patients, highlighting the potential role of astrocyte-mediated inflammatory signaling in BD neuropathology.


Subject(s)
Astrocytes/pathology , Bipolar Disorder/pathology , Inflammation/pathology , Neurons/pathology , Coculture Techniques , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Interleukin-1beta/pharmacology , Interleukin-6/metabolism , Neuroglia/drug effects , Neuroglia/pathology , Neurons/drug effects , Neurons/metabolism
14.
Article in English | MEDLINE | ID: mdl-31767646

ABSTRACT

Brain disorders, from neurodegenerative to psychiatric disorders, are among the most challenging conditions to study because of the intricate nature of the human brain and the limitations of existing model systems in recapitulating all these intricacies. However, innovations in stem cell technologies now allow us to reprogram patient somatic cells to induced pluripotent stem cells (iPSCs), which can then be differentiated to disease-relevant neural and glial cells. iPSCs are a valuable tool to model brain disorders, as they can be derived from patients with known symptom histories, genetics, and drug-response profiles. Here, we discuss the premise and validity of the iPSC-based in vitro model system and highlight key findings from the most commonly studied neurodegenerative and psychiatric disorders.


Subject(s)
Brain Diseases/metabolism , Brain/physiopathology , Induced Pluripotent Stem Cells/metabolism , Animals , Brain/physiology , Cell Differentiation , Central Nervous System , Chemistry, Pharmaceutical/methods , Drug Design , Genetic Techniques , Humans , Models, Biological , Models, Neurological , Neurodegenerative Diseases , Neuroglia/metabolism , Neurons/metabolism , Pluripotent Stem Cells/physiology , Stem Cell Transplantation
15.
Focus (Am Psychiatr Publ) ; 17(3): 294-307, 2019 Jul.
Article in English | MEDLINE | ID: mdl-32015721

ABSTRACT

Reprinted with permission from Open Biol. 8: 180031. The Royal Society.

16.
Antioxid Redox Signal ; 31(4): 275-317, 2019 08 01.
Article in English | MEDLINE | ID: mdl-30585734

ABSTRACT

Significance: Our current knowledge of the pathophysiology and molecular mechanisms causing psychiatric disorders is modest, but genetic susceptibility and environmental factors are central to the etiology of these conditions. Autism, schizophrenia, bipolar disorder and major depressive disorder show genetic gene risk overlap and share symptoms and metabolic comorbidities. The identification of such common features may provide insights into the development of these disorders. Recent Advances: Multiple pieces of evidence suggest that brain energy metabolism, mitochondrial functions and redox balance are impaired to various degrees in psychiatric disorders. Since mitochondrial metabolism and redox signaling can integrate genetic and environmental environmental factors affecting the brain, it is possible that they are implicated in the etiology and progression of psychiatric disorders. Critical Issue: Evidence for direct links between cellular mitochondrial dysfunction and disease features are missing. Future Directions: A better understanding of the mitochondrial biology and its intracellular connections to the nuclear genome, the endoplasmic reticulum and signaling pathways, as well as its role in intercellular communication in the organism, is still needed. This review focuses on the findings that implicate mitochondrial dysfunction, the resultant metabolic changes and oxidative stress as important etiological factors in the context of psychiatric disorders. We also propose a model where specific pathophysiologies of psychiatric disorders depend on circuit-specific impairments of mitochondrial dysfunction and redox signaling at specific developmental stages.


Subject(s)
Mental Disorders/metabolism , Mitochondria/metabolism , Animals , Humans , Oxidation-Reduction
17.
Elife ; 82019 02 07.
Article in English | MEDLINE | ID: mdl-30730291

ABSTRACT

Comparative analyses of neuronal phenotypes in closely related species can shed light on neuronal changes occurring during evolution. The study of post-mortem brains of nonhuman primates (NHPs) has been limited and often does not recapitulate important species-specific developmental hallmarks. We utilize induced pluripotent stem cell (iPSC) technology to investigate the development of cortical pyramidal neurons following migration and maturation of cells grafted in the developing mouse cortex. Our results show differential migration patterns in human neural progenitor cells compared to those of chimpanzees and bonobos both in vitro and in vivo, suggesting heterochronic changes in human neurons. The strategy proposed here lays the groundwork for further comparative analyses between humans and NHPs and opens new avenues for understanding the differences in the neural underpinnings of cognition and neurological disease susceptibility between species.


Subject(s)
Neurons/cytology , Pan paniscus/physiology , Pan troglodytes/physiology , Animals , Cell Differentiation , Cell Line , Cell Movement/genetics , Dendrites/metabolism , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Species Specificity
18.
Genome Med ; 10(1): 1, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29301565

ABSTRACT

Our understanding of the neurobiology of psychiatric disorders remains limited, and biomarker-based clinical management is yet to be developed. Induced pluripotent stem cell (iPSC) technology has revolutionized our capacity to generate patient-derived neurons to model psychiatric disorders. Here, we highlight advantages and caveats of iPSC disease modeling and outline strategies for addressing current challenges.


Subject(s)
Induced Pluripotent Stem Cells/pathology , Mental Disorders/pathology , Models, Biological , Neurons/pathology , Animals , Humans , Mental Disorders/genetics
19.
Open Biol ; 8(5)2018 05.
Article in English | MEDLINE | ID: mdl-29794033

ABSTRACT

Personalized medicine has become increasingly relevant to many medical fields, promising more efficient drug therapies and earlier intervention. The development of personalized medicine is coupled with the identification of biomarkers and classification algorithms that help predict the responses of different patients to different drugs. In the last 10 years, the Food and Drug Administration (FDA) has approved several genetically pre-screened drugs labelled as pharmacogenomics in the fields of oncology, pulmonary medicine, gastroenterology, haematology, neurology, rheumatology and even psychiatry. Clinicians have long cautioned that what may appear to be similar patient-reported symptoms may actually arise from different biological causes. With growing populations being diagnosed with different psychiatric conditions, it is critical for scientists and clinicians to develop precision medication tailored to individual conditions. Genome-wide association studies have highlighted the complicated nature of psychiatric disorders such as schizophrenia, bipolar disorder, major depression and autism spectrum disorder. Following these studies, association studies are needed to look for genomic markers of responsiveness to available drugs of individual patients within the population of a specific disorder. In addition to GWAS, the advent of new technologies such as brain imaging, cell reprogramming, sequencing and gene editing has given us the opportunity to look for more biomarkers that characterize a therapeutic response to a drug and to use all these biomarkers for determining treatment options. In this review, we discuss studies that were performed to find biomarkers of responsiveness to different available drugs for four brain disorders: bipolar disorder, schizophrenia, major depression and autism spectrum disorder. We provide recommendations for using an integrated method that will use available techniques for a better prediction of the most suitable drug.


Subject(s)
Antipsychotic Agents/therapeutic use , Genetic Markers , Mental Disorders/drug therapy , Pharmacogenomic Variants , Antipsychotic Agents/pharmacology , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/genetics , Bipolar Disorder/drug therapy , Bipolar Disorder/genetics , Clinical Trials as Topic , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/genetics , Humans , Mental Disorders/genetics , Pharmacogenetics , Precision Medicine , Schizophrenia/drug therapy , Schizophrenia/genetics
20.
Neuropsychopharmacology ; 32(7): 1504-19, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17164818

ABSTRACT

Stress regulation of brain-derived neurotrophic factor (BDNF) is implicated in the hippocampal damage observed in depression. BDNF has a complex gene structure with four 5' untranslated exons (I-IV) with unique promoters, and a common 3' coding exon (V). To better understand the stress regulation of BDNF, we addressed whether distinct stressors differentially regulate exon-specific BDNF transcripts in the postnatal and adult hippocampus. The early life stress of maternal separation (MS) resulted in a time point-dependent differential upregulation of BDNF transcripts restricted to early postnatal life (P14-BDNF II, P21-BDNF IV, V). In adulthood, distinct stressors regulated BDNF transcripts in a signature manner. Immobilization stress, administered once, decreased all BDNF splice variants but had differing effects on BDNF I/II (increase) and III/IV (decrease) when administered chronically. Although immobilization stress reduced BDNF (V) mRNA, chronic unpredictable stress did not influence total BDNF despite altering specific BDNF transcripts. Furthermore, a prior history of MS altered the signature pattern in which adult-onset stress regulated specific BDNF transcripts. We also examined the expression of cyclic AMP response element-binding protein (CREB), an upstream transcriptional activator of BDNF, and observed a CREB induction in the postnatal hippocampus following MS. As a possible consequence of enhanced CREB and BDNF expression following MS, we examined hippocampal progenitor proliferation and observed a significant increase restricted to early life. These results suggest that alterations in CREB/BDNF may contribute to the generation of individual differences in stress neurocircuitry, providing a substrate for altered vulnerability to depressive disorders.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Gene Expression Regulation, Developmental/genetics , Hippocampus/growth & development , Hippocampus/metabolism , Stress, Psychological/metabolism , Aging/physiology , Animals , Animals, Newborn , Cell Differentiation/physiology , Male , Maternal Deprivation , Neural Pathways/growth & development , Neural Pathways/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Stem Cells/metabolism , Stress, Psychological/physiopathology , Transcriptional Activation/physiology , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL