Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Cells ; 13(5)2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38474374

ABSTRACT

Mitochondria, the energy suppliers of the cells, play a central role in a variety of cellular processes essential for survival or leading to cell death. Consequently, mitochondrial dysfunction is implicated in numerous general and CNS disorders. The clinical manifestations of mitochondrial dysfunction include metabolic disorders, dysfunction of the immune system, tumorigenesis, and neuronal and behavioral abnormalities. In this review, we focus on the mitochondrial role in the CNS, which has unique characteristics and is therefore highly dependent on the mitochondria. First, we review the role of mitochondria in neuronal development, synaptogenesis, plasticity, and behavior as well as their adaptation to the intricate connections between the different cell types in the brain. Then, we review the sparse knowledge of the mechanisms of exogenous mitochondrial uptake and describe attempts to determine their half-life and transplantation long-term effects on neuronal sprouting, cellular proteome, and behavior. We further discuss the potential of mitochondrial transplantation to serve as a tool to study the causal link between mitochondria and neuronal activity and behavior. Next, we describe mitochondrial transplantation's therapeutic potential in various CNS disorders. Finally, we discuss the basic and reverse-translation challenges of this approach that currently hinder the clinical use of mitochondrial transplantation.


Subject(s)
Central Nervous System Diseases , Mitochondrial Diseases , Humans , Mitochondria/metabolism , Central Nervous System/metabolism , Brain/metabolism , Central Nervous System Diseases/metabolism , Mitochondrial Diseases/metabolism
2.
Neurosci Res ; 192: 83-92, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36717018

ABSTRACT

Mitochondrial dysfunction was shown to be involved in schizophrenia pathophysiology. Abnormal energy states can lead to alterations in neural function and thereby to the cognitive and behavioral aberrations characteristics of schizophrenia. Voltage-dependent anion-selective channels (VDAC) are located in the outer mitochondrial membrane and are involved in mitochondrial energy production. Only few studies explored VDAC genes' expression in schizophrenia, and their results were not consistent. We conducted a systematic meta-analysis of ten brain samples gene expression datasets (overall 368 samples, 179 schizophrenia, 189 controls). In addition, we conducted a meta-analysis of three blood samples datasets (overall 300 samples, 167 schizophrenia, 133 controls). Pairwise correlation analysis was conducted between the VDAC and proteasome subunit genes' expression patterns. VDAC1, VDAC2 and VDAC3 showed significant down-regulation in brain samples of patients with schizophrenia. They also showed significant positive correlations with the proteasome subunit genes' expression levels. Our findings suggest that VDAC genes might play a role in mitochondrial dysfunction in schizophrenia. VDAC1 was down-regulated also in blood samples, which suggests its potential role as a biomarker for schizophrenia. The correlation with proteasome subunits, which were previously shown to be down-regulated in a subgroup of the patients, suggests that our findings might characterize a subgroup of the patients. This direction has the potential to lead to patients' stratification and more precisely-targeted therapy and necessitates further study.


Subject(s)
Proteasome Endopeptidase Complex , Schizophrenia , Humans , Down-Regulation , Proteasome Endopeptidase Complex/genetics , Schizophrenia/genetics , Protein Isoforms/genetics , Gene Expression , Brain
3.
J Psychiatr Res ; 158: 350-359, 2023 02.
Article in English | MEDLINE | ID: mdl-36640659

ABSTRACT

Schizophrenia is a chronic and debilitating mental disorder, with unknown pathophysiology. Converging lines of evidence suggest that mitochondrial functioning may be compromised in schizophrenia. Postmortem brain samples of individuals with schizophrenia showed dysregulated expression levels of genes encoding enzyme complexes comprising the mitochondrial electron transport chain (ETC), including ATP synthase, the fifth ETC complex. However, there are inconsistencies regarding the direction of change, i.e., up- or down-regulation, and differences between female and male patients were hardly examined. We have performed a systematic meta-analysis of the expression of 16 ATP synthase encoding genes in postmortem brain samples of individuals with schizophrenia vs. healthy controls of three regions: Brodmann Area 10 (BA10), BA22/Superior Temporal Gyrus (STG) and the cerebellum. Eight independent datasets were integrated (overall 294brain samples, 145 of individuals with schizophrenia and 149 controls). The meta-analysis was applied to all individuals with schizophrenia vs. the controls, and also to female and male patients vs. age-matched controls, separately. A significant down-regulation of two ATP synthase encoding genes was detected in schizophrenia, ATP5A1 and ATP5H, and a trend towards down-regulation of five further ATP synthase genes. The down-regulation tendency was shown for both females and males with schizophrenia. Our findings support the hypothesis that schizophrenia is associated with reduced ATP synthesis via the oxidative phosphorylation system, which is caused by reduced cellular demand of ATP. Abnormal cellular energy metabolism can lead to alterations in neural function and brain circuitry, and thereby to the cognitive and behavioral aberrations characteristic of schizophrenia.


Subject(s)
Brain , Mitochondrial Proton-Translocating ATPases , Schizophrenia , Female , Humans , Male , Adenosine Triphosphate/metabolism , Brain/metabolism , Brain/pathology , Down-Regulation , Schizophrenia/metabolism , Temporal Lobe/metabolism , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism
4.
Mol Psychiatry ; 28(3): 1170-1181, 2023 03.
Article in English | MEDLINE | ID: mdl-36380234

ABSTRACT

Ample evidence implicate mitochondria in early brain development. However, to the best of our knowledge, there is only circumstantial data for mitochondria involvement in late brain development occurring through adolescence, a critical period in the pathogenesis of various psychiatric disorders, specifically schizophrenia. In schizophrenia, neurodevelopmental abnormalities and mitochondrial dysfunction has been repeatedly reported. Here we show a causal link between mitochondrial transplantation in adolescence and brain functioning in adulthood. We show that transplantation of allogenic healthy mitochondria into the medial prefrontal cortex of adolescent rats was beneficial in a rat model of schizophrenia, while detrimental in healthy control rats. Specifically, disparate initial changes in mitochondrial function and inflammatory response were associated with opposite long-lasting changes in proteome, neurotransmitter turnover, neuronal sprouting and behavior in adulthood. A similar inverse shift in mitochondrial function was also observed in human lymphoblastoid cells deived from schizophrenia patients and healthy subjects due to the interference of the transplanted mitochondria with their intrinsic mitochondrial state. This study provides fundamental insights into the essential role of adolescent mitochondrial homeostasis in the development of normal functioning adult brain. In addition, it supports a therapeutic potential for mitochondria manipulation in adolescence in disorders with neurodevelopmental and bioenergetic deficits, such as schizophrenia, yet emphasizes the need to monitor individuals' state including their mitochondrial function and immune response, prior to intervention.


Subject(s)
Schizophrenia , Adult , Rats , Humans , Animals , Adolescent , Mitochondria , Brain , Neurons , Disease Models, Animal
5.
Methods Mol Biol ; 2277: 187-201, 2021.
Article in English | MEDLINE | ID: mdl-34080153

ABSTRACT

Mitochondria, similar to living cells and organelles, have a negative membrane potential, which ranges between (-108) and (150) mV as compared to (-70) and (-90) mV of the plasma membrane. Therefore, permeable lipophilic cations tend to accumulate in the mitochondria. Those cations which exhibit fluorescence activity after accumulation into energized systems are widely used to decipher changes in membrane potential by imaging techniques. Here we describe the use of two different dyes for labeling mitochondrial membrane potential (Δψm) in live cells. One is the lipophilic cation 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazol-carbocyanine iodide (JC-1), which alters reversibly its color from green (J-monomer, at its low concentration in the cytosol) to red (J-aggregates, at its high concentration in active mitochondria) with increasing mitochondrial membrane potential (Δψm). The other is MitoTracker® Orange, a mitochondrion-selective probe which passively diffuses across the plasma membrane and accumulates in active mitochondria depending on their Δψm. We show that in addition to changes in Δψm, these specific dyes can be used to follow alterations in mitochondrial distribution and mitochondrial network connectivity. We suggest that JC-1 is a preferable probe to compare between different cell types and cell state, as a red to green ratio of fluorescence intensities is used for analysis. This ratio depends only on the mitochondrial membrane potential and not on other cellular and/or mitochondrial-dependent or independent factors that may alter, for example, due to treatment or disease state. However, in cells labeled either with green or red fluorescence protein, JC-1 cannot be used. Therefore, other dyes are preferable. We demonstrate various applications of JC-1 and MitoTracker Orange staining to study mitochondrial abnormalities in different cell types derived from schizophrenia patients and healthy subjects.


Subject(s)
Image Processing, Computer-Assisted/methods , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Schizophrenia/metabolism , Benzimidazoles/chemistry , Carbocyanines/chemistry , Cell Culture Techniques , Fibroblasts/metabolism , Fibroblasts/pathology , Fluorescent Dyes/chemistry , Humans , Microscopy, Confocal/instrumentation , Microscopy, Confocal/methods , Mitochondria/pathology , Proof of Concept Study , Schizophrenia/pathology , Xanthenes/chemistry
6.
Sci Rep ; 10(1): 12258, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32703977

ABSTRACT

Pharmacological treatment of mental disorders is currently decided based on "trial and error" strategy. Mitochondrial multifaceted dysfunction is assumed to be a major factor in the pathophysiology and treatment of schizophrenia (SZ) and bipolar disorder (BD). This study aimed to explore the feasibility of using a profile of mitochondrial function parameters as a tool to predict the optimal drug for an individual patient (personalized medicine). Healthy controls (n = 40), SZ (n = 48) and BD (n = 27) patients were recruited. Mental and global state of the subjects, six mitochondrial respiration parameters and 14 mitochondrial function-related proteins were assessed in fresh lymphocytes following in-vitro or in-vivo treatment with five antipsychotic drugs and two mood-stabilizers. In healthy controls, hierarchal clustering shows a drug-specific effect profile on the different mitochondrial parameters following in-vitro exposure. Similar changes were observed in untreated SZ and BD patients with psychosis. Following a month of treatment of the latter patients, only responders showed a significant correlation between drug-induced in-vitro effect (prior to in-vivo treatment) and short-term in-vivo treatment effect for 45% of the parameters. Long- but not short-term psychotropic treatment normalized mitochondria-related parameters in patients with psychosis. Taken together, these data substantiate mitochondria as a target for psychotropic drugs and provide a proof of concept for selective mitochondrial function-related parameters as a predictive tool for an optimized psychotropic treatment in a given patient. This, however, needs to be repeated with an expanded sample size and additional mitochondria related parameters.


Subject(s)
Antipsychotic Agents/pharmacology , Bipolar Disorder/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Psychotic Disorders/metabolism , Adolescent , Adult , Antipsychotic Agents/therapeutic use , Biomarkers , Bipolar Disorder/drug therapy , Bipolar Disorder/etiology , Case-Control Studies , Female , Gene Expression , Humans , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Middle Aged , Prognosis , Proof of Concept Study , Psychotic Disorders/drug therapy , Psychotic Disorders/etiology , Psychotropic Drugs/pharmacology , Psychotropic Drugs/therapeutic use , Young Adult
7.
Mol Psychiatry ; 25(4): 805-820, 2020 04.
Article in English | MEDLINE | ID: mdl-30531937

ABSTRACT

Mitochondria together with other cellular components maintain a constant crosstalk, modulating transcriptional and posttranslational processes. We and others demonstrated mitochondrial multifaceted dysfunction in schizophrenia, with aberrant complex I (CoI) as a major cause. Here we show deficits in CoI activity and homeostasis in schizophrenia-derived cell lines. Focusing on a core CoI subunit, NDUFV2, one of the most severely affected subunits in schizophrenia, we observed reduced protein level and functioning, with no change in mRNA transcripts. We further show that NDUFV2 pseudogene (NDUFV2P1) expression is increased in schizophrenia-derived cells and in postmortem brain specimens. In schizophrenia and controls pooled samples, NDUFV2P1 level demonstrated a significant inverse correlation with NDUFV2 pre- and matured protein level and with CoI-driven cellular respiration. Our data suggest a role for a pseudogene in its parent-gene regulation and possibly in CoI dysfunction in schizophrenia. The abnormal expression of the pseudogene may be one element of a vicious circle in which CoI deficits lead to mitochondrial dysfunction potentially affecting genome-wide regulation of gene expression, including the expression of pseudogenes.


Subject(s)
Electron Transport Complex I/genetics , NADH Dehydrogenase/genetics , Schizophrenia/genetics , Electron Transport Complex I/metabolism , Gene Expression , Humans , Mitochondria/metabolism , NADH Dehydrogenase/metabolism , Pseudogenes , RNA, Messenger/metabolism , Schizophrenia/metabolism
8.
J Neural Transm (Vienna) ; 127(2): 159-168, 2020 02.
Article in English | MEDLINE | ID: mdl-31848775

ABSTRACT

Parkinson's disease (PD) and schizophrenia (SZ) are two CNS disorders in which dysfunctions in the dopaminergic system and mitochondria are major pathologies. The symptomology of both, PD a neurodegenerative disorder and SZ a neurodevelopmental disorder, is completely different. However, the pharmacological treatment of each of the diseases can cause a shift of symptoms into those characteristic of the other disease. In this review, I describe a pathological interaction between dopamine and mitochondria in both disorders, which due to differences in the extent of oxidative stress leads either to cell death and tissue degeneration as in PD substantia nigra pars compacta or to distorted neuronal activity, imbalanced neuronal circuitry and abnormal behavior and cognition in SZ. This review is in the honor of Moussa Youdim who introduced me to the secrets of research work. His enthusiasm, curiosity and novelty-seeking inspired me throughout my career. Thank you Moussa.


Subject(s)
Dopamine , Mitochondria , Neurons , Parkinson Disease , Schizophrenia , Animals , Dopamine/metabolism , Humans , Mitochondria/metabolism , Neurons/metabolism , Neurons/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Schizophrenia/metabolism , Schizophrenia/physiopathology
9.
Eur Neuropsychopharmacol ; 29(5): 577-589, 2019 05.
Article in English | MEDLINE | ID: mdl-30948194

ABSTRACT

Accumulating data point to heme involvement in neuropsychiatric disorders. Heme plays a role in major cellular processes such as signal transduction, protein complex assembly and regulation of transcription and translation. Its synthesis involves the mitochondria, which dysfunction, specifically that of the complex I (Co-I) of the electron transport chain is involved in the pathophysiology of schizophrenia (SZ). Here we aimed to demonstrate that deficits in Co-I affect heme metabolism. We show a significant decrease in heme levels in Co-I deficient SZ-derived EBV transformed lymphocytes (lymphoblastoid cell lines - LCLs) as compared to healthy subjects-derived cells (n = 9/cohort). Moreover, protein levels assessed by immunoblotting and mRNA levels assessed by qRT-PCR of heme catabolic enzyme, heme Oxygenase 1 (HO-1), and protein levels of heme downstream target phosphorylated eukaryotic initiation factor 2-alpha (Peif2a/eif2a) were significantly elevated in SZ-derived cells. In contrast, protein and mRNA levels of heme synthesis rate limiting enzyme aminolevulinic acid synthase-1 (ALAS1) were unchanged in SZ derived LCLs. In addition, inhibition of Co-I by rotenone in healthy subjects-derived LCLs (n = 4/cohort) exhibited an initial increase followed by a later decrease in heme levels. These findings were associated with opposite changes in heme's downstream target and HO-1 level, similar to our findings in SZ-derived cells. We also show a brain region specific pattern of impairment in Co-I subunits and in HO-1 and PeIF2α/eIF2α in the Poly-IC rat model of SZ (n = 6/cohort). Our results provide evidence for a link between CoI and heme metabolism both in-vitro and in-vivo suggesting its contribution to SZ pathophysiology.


Subject(s)
Electron Transport Complex I/metabolism , Hemeproteins/metabolism , Mitochondria/metabolism , Schizophrenia/metabolism , Adult , Animals , Cell Line , Female , Humans , Interferon Inducers/toxicity , Male , Middle Aged , Poly I-C/toxicity , Rats , Schizophrenia/chemically induced , Young Adult
10.
Front Cell Neurosci ; 12: 321, 2018.
Article in English | MEDLINE | ID: mdl-30319360

ABSTRACT

In the last decade, there is an increasing application of induced pluripotent stem cells (iPSCs) for disease modeling. The iPSC technology enables the study of patient-specific neuronal cell lines in vitro to evaluate dysfunction at the cellular level and identify the responsible genetic factors. This approach might be particularly valuable for filling the gap of knowledge at the cellular and molecular levels underlying the pathophysiology of various neurodevelopmental and/or psychiatric disorders, such as attention-deficit hyperactivity disorder (ADHD). However, the invasiveness of skin biopsy or blood withdrawal might represent a major impediment in such protected population. Using hair derived keratinocytes as starting somatic cells circumvents this problem as sample collections can be performed non-invasively. Here we describe an improved, convenient, standardized and effective method to culture and reprogram hair derived keratinocytes from three healthy controls and one ADHD patient into iPSCs, which in turn will be used to generate differentiated neuronal cells. All the cell types were maintained in highly defined, serum-free conditions and showed expression of the respective key marker genes, assessed by both immunocytochemistry and qRT-PCR. The described in vitro personalized neuronal model has its advantage in modeling neurodevelopmental trajectories since it can recapitulate key processes of brain development at the cellular and molecular level and is intended to be used as for example studying ADHD etiopathology.

11.
Clin Exp Pharmacol Physiol ; 45(10): 1028-1037, 2018 10.
Article in English | MEDLINE | ID: mdl-29851136

ABSTRACT

Chemotherapy-induced cognitive changes is a major burden on a substantial number of cancer survivors. The mechanism of this sequel is unknown. In this study, we followed long-term effects of early in life mithramycin (MTR) treatment on behaviour and on the normal course of alterations of gene expression in brain. Between post-natal days (PND) 7 and 10, male rats were divided into 2 groups, 1 receiving MTR (0.1 mg/kg s.c. per day) and the other receiving saline. At PND11, frontal cortex tissue samples were dissected from 4 rats from each group. At PND 65 the remaining rats underwent behavioural tests after which all the rats were decapitated and their prefrontal cortex incised. Rats treated transiently with MTR early in life, showed impairments in spatial working memory and anxious-like behaviour in adulthood. The immediate molecular effect of MTR was expressed in a limited number of altered genes of different unconnected trajectories, which were simultaneously distorted by the drug. In contrast, 3 months later we observed a change in the expression of more than 1000 genes that converged into specific cellular processes. Time-dependent gene expression dynamics of several genes was significantly different between treated and untreated rats. The differences in the total number of altered genes and in gene expression trends, immediately and long after MTR treatment cessation, suggest the evolution of a new cellular homeostatic set point, which can lead to behavioural abnormalities following chemotherapy treatment.


Subject(s)
Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/genetics , Gene Expression Regulation/drug effects , Plicamycin/adverse effects , Animals , Anxiety/complications , Cognitive Dysfunction/complications , Cognitive Dysfunction/physiopathology , Male , Memory, Short-Term/drug effects , Phenotype , Rats , Spatial Memory/drug effects
12.
Biol Psychiatry ; 83(9): 770-779, 2018 05 01.
Article in English | MEDLINE | ID: mdl-28965983

ABSTRACT

The neurobiology of psychiatric disorders is still unclear, although changes in multiple neuronal systems, specifically the dopaminergic, glutamatergic, and gamma-aminobutyric acidergic systems as well as abnormalities in synaptic plasticity and neural connectivity, are currently suggested to underlie their pathophysiology. A growing body of evidence suggests multifaceted mitochondrial dysfunction in mental disorders, which is in line with their role in neuronal activity, growth, development, and plasticity. In this review, we describe the main endeavors toward development of treatments that will enhance mitochondrial function and their transition into clinical use in congenital mitochondrial diseases and chronic disorders such as types 1 and 2 diabetes, cardiovascular disorders, and cancer. In addition, we discuss the relevance of mitochondrial targeted treatments to mental disorders and their potential to become a novel therapeutic strategy that will improve the efficiency of the current treatments.


Subject(s)
Antioxidants/therapeutic use , Mental Disorders/therapy , Mitochondria , Mitochondrial Diseases/therapy , Humans , Mental Disorders/etiology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/transplantation , Mitochondrial Diseases/complications
13.
Schizophr Bull ; 44(2): 432-442, 2018 02 15.
Article in English | MEDLINE | ID: mdl-28586483

ABSTRACT

Dysfunction of mitochondria, key players in various essential cell processes, has been repeatedly reported in schizophrenia (SZ). Recently, several studies have reported functional recovery and cellular viability following mitochondrial transplantation, mostly in ischemia experimental models. Here, we aimed to demonstrate beneficial effects of isolated active normal mitochondria (IAN-MIT) transfer in vitro and in vivo, using SZ-derived induced pluripotent stem cells (iPSCs) differentiating into glutamatergic neuron, as well as a rodent model of SZ. First, we show that IAN-MIT enter various cell types without manipulation. Next, we show that IAN-MIT transfer into SZ-derived lymphoblasts induces long-lasting improvement in various mitochondrial functions including cellular oxygen consumption and mitochondrial membrane potential (Δ ψ m). We also demonstrate improved differentiation of SZ-derived iPSCs into neurons, by increased expression of neuronal and glutamatergic markers ß3-tubulin, synapsin1, and Tbr1 and by an activation of the glutamate-glutamine cycle. In the animal model, we show that intra-prefrontal cortex injection of IAN-MIT in adolescent rats exposed prenatally to a viral mimic prevents mitochondrial Δ ψ m and attentional deficit at adulthood. Our results provide evidence for a direct link between mitochondrial function and SZ-related deficits both in vitro and in vivo and suggest a therapeutic potential for IAN-MIT transfer in diseases with bioenergetic and neurodevelopmental abnormalities such as SZ.


Subject(s)
Cell Differentiation/physiology , Cognitive Dysfunction , Induced Pluripotent Stem Cells/metabolism , Mitochondria , Neurons/metabolism , Prefrontal Cortex , Schizophrenia , Animals , Attention/physiology , Behavior, Animal/physiology , Cells, Cultured , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/therapy , Disease Models, Animal , Female , Humans , Male , Mitochondria/metabolism , Mitochondria/transplantation , Rats , Rats, Wistar , Schizophrenia/metabolism , Schizophrenia/therapy
14.
Schizophr Res ; 187: 3-10, 2017 09.
Article in English | MEDLINE | ID: mdl-27802911

ABSTRACT

Mitochondria are key players in various essential cellular processes beyond being the main energy supplier of the cell. Accordingly, they are involved in neuronal synaptic transmission, neuronal growth and sprouting and consequently neuronal plasticity and connectivity. In addition, mitochondria participate in the modulation of gene transcription and inflammation as well in physiological responses in health and disease. Schizophrenia is currently regarded as a neurodevelopmental disorder associated with impaired immune system, aberrant neuronal differentiation and abnormalities in various neurotransmitter systems mainly the dopaminergic, glutaminergic and GABAergic. Ample evidence has been accumulated over the last decade indicating a multifaceted dysfunction of mitochondria in schizophrenia. Indeed, mitochondrial deficit can be of relevance for the majority of the pathologies observed in this disease. In the present article, we overview specific deficits of the mitochondria in schizophrenia, with a focus on the first complex (complex I) of the mitochondrial electron transport chain (ETC). We argue that complex I, being a major factor in the regulation of mitochondrial ETC, is a possible key modulator of various functions of the mitochondria. We review biochemical, molecular, cellular and functional evidence for mitochondrial impairments and their possible convergence to impact in-vitro neuronal differentiation efficiency in schizophrenia. Mitochondrial function in schizophrenia may advance our knowledge of the disease pathophysiology and open the road for new treatment targets for the benefit of the patients.


Subject(s)
Electron Transport Complex I/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Schizophrenia/metabolism , Animals , Antipsychotic Agents/pharmacology , Humans , Mitochondria/drug effects , Mitochondrial Diseases/drug therapy , Schizophrenia/drug therapy
15.
World J Psychiatry ; 6(3): 294-302, 2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27679768

ABSTRACT

It is generally assumed that behavior results from an interaction between susceptible genes and environmental stimuli during critical life stages. The present article reviews the main theoretical and practical concepts in the research of gene environment interaction, emphasizing the need for models simulating real life complexity. We review a novel approach to study gene environment interaction in which a brief post-natal interference with the expression of multiple genes, by hindering the activity of the ubiquitous transcription factor specificity protein 1 (Sp1) is followed by later-in-life exposure of rats to stress. Finally, this review discusses the role of peripheral processes in behavioral responses, with the Sp1 model as one example demonstrating how specific behavioral patterns are linked to modulations in both peripheral and central physiological processes. We suggest that models, which take into account the tripartite reciprocal interaction between the central nervous system, peripheral systems and environmental stimuli will advance our understanding of the complexity of behavior.

16.
Can J Psychiatry ; 61(8): 457-69, 2016 08.
Article in English | MEDLINE | ID: mdl-27412728

ABSTRACT

Mitochondria are key players in the generation and regulation of cellular bioenergetics, producing the majority of adenosine triphosphate molecules by the oxidative phosphorylation system (OXPHOS). Linked to numerous signaling pathways and cellular functions, mitochondria, and OXPHOS in particular, are involved in neuronal development, connectivity, plasticity, and differentiation. Impairments in a variety of mitochondrial functions have been described in different general and psychiatric disorders, including schizophrenia (SCZ), a severe, chronic, debilitating illness that heavily affects the lives of patients and their families. This article reviews findings emphasizing the role of OXPHOS in the pathophysiology of SCZ. Evidence accumulated during the past few decades from imaging, transcriptomic, proteomic, and metabolomic studies points at OXPHOS deficit involvement in SCZ. Abnormalities have been reported in high-energy phosphates generated by the OXPHOS, in the activity of its complexes and gene expression, primarily of complex I (CoI). In addition, cellular signaling such as cAMP/protein kinase A (PKA) and Ca(+2), neuronal development, connectivity, and plasticity have been linked to OXPHOS function and are reported to be impaired in SCZ. Finally, CoI has been shown as a site of interaction for both dopamine (DA) and antipsychotic drugs, further substantiating its role in the pathology of SCZ. Understanding the role of mitochondria and the OXPHOS in particular may encourage new insights into the pathophysiology and etiology of this debilitating disorder.


Subject(s)
Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Oxidative Phosphorylation , Schizophrenia/metabolism , Signal Transduction/physiology , Humans
17.
Eur Neuropsychopharmacol ; 25(10): 1695-705, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26271721

ABSTRACT

Previously, we showed that a transient early-in-life interference with the expression of multiple genes by mithramycin (MTR) followed by later-in-life exposure to chronic stress, leads to a "daring" and novelty seeking behavior in rats. In this study we searched for molecular changes that contribute to this behavioral alteration. We applied a non-hypothesis driven strategy using whole genome cDNA array analysis (WGA) followed by Genome Scale Metabolic modeling analysis (GSMM). Gene expression validation was performed by qRT-PCR and immunoblotting. Brain and serum amino acids levels were measured by HPLC. WGA data directed us towards metabolic pathways and GSMM pointed at branched chain amino acids (BCAA) pathway. Out of 21 amino acids analyzed in the prefrontal cortex of MTR+Stress rats only tryptophan, whose brain levels depend on serum BCAA levels, showed a significant decrease. No change was observed in serotonin or kynurenine levels. However, a significant reduction in mRNA and protein levels of the large neutral amino acid transporter (LAT1), which transports BCAA and tryptophan into the brain, as well as in serum levels of tryptophan/BCAA ratio were observed. The latter may be attributed to the failure to increase serum insulin, following stress, in rats pre-exposed to mithramycin. Finally, significant correlations were observed between the anxiety index and tryptophan and between T-maze errors and LAT1. This study shows a specific behavioral pattern, which is linked to modulations in fluxes of amino acids both peripheral and central, which converge and reciprocally interact, and may thus be equally important targets for therapeutic intervention.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Anxiety/metabolism , Maze Learning/physiology , Prefrontal Cortex/metabolism , Stress, Psychological/metabolism , Tryptophan/metabolism , Animals , Blood Glucose/metabolism , Disease Models, Animal , Exploratory Behavior/physiology , Insulin/blood , Kynurenine/metabolism , Large Neutral Amino Acid-Transporter 1/metabolism , Male , Plicamycin/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/growth & development , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/metabolism , Random Allocation , Rats, Wistar , Serotonin/metabolism
18.
Methods Mol Biol ; 1265: 425-39, 2015.
Article in English | MEDLINE | ID: mdl-25634294

ABSTRACT

Mitochondria, similar to living cells and organelles, have negative membrane potential and can therefore accumulate permeable lipophilic cations. Those cations which exhibit fluorescence activity after accumulation into energized systems are widely used to decipher changes in membrane potential by imaging techniques. Here we describe the use of the lipophilic cation 5,5',6,6'tetrachloro-1,1',3,3'-tetraethylbenzimidazol-carbocyanine iodide (JC-1), which alters reversibly its color from green (J-monomer, at its low concentration in the cytosol) to red (J-aggregates, at its high concentration in active mitochondria) with increasing mitochondrial membrane potential (Δψm). We show that in addition to changes in Δψm, this specific dye can be used to follow alterations in mitochondrial distribution and mitochondrial network connectivity. We suggest that JC-1 is a preferable probe to compare between treatment groups, as the ratio of green to red fluorescence intensities is used for analysis. This ratio depends only on the mitochondrial membrane potential and not on other mitochondrial dependent or independent factors. We demonstrate various applications of JC-1 staining to study mitochondrial abnormalities in different cell types derived from schizophrenia patients and healthy subjects.


Subject(s)
Microscopy, Fluorescence , Mitochondria/metabolism , Molecular Imaging/methods , Schizophrenia/metabolism , Biological Transport , Cell Line , Humans , Image Processing, Computer-Assisted , Membrane Potential, Mitochondrial , Microscopy, Fluorescence/methods
19.
Int J Neuropsychopharmacol ; 17(2): 337-40, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24229565

ABSTRACT

Negative symptoms in schizophrenia are associated with decreased dopaminergic activity in the prefrontal cortex (PFC). It is hypothesized that increasing dopamine levels would alleviate negative symptoms. Termination of dopamine activity in the PFC is mainly via catechol-O-methyl tranferase (COMT) activity. Hence, inhibition of COMT activity with entacapone should reverse PFC dopaminergic transmission. To assess the efficacy of entacapone addition to antipsychotic treatment in patients with residual schizophrenia, we conducted a double-blind, randomised, placebo-controlled study for 12 wk of treatment with entacapone or placebo. Clinical measures (PANSS, CGI and QLS) were obtained at baseline and at weeks 4, 8 and 12 and cognitive functions were assessed by the RBANSS. Significant improvement over time in PANSS and QLS scores was observed in both groups. However, entacapone did not demonstrate a beneficial effect compared to placebo. Therefore, this study does not support a therapeutic role for entacapone in residual schizophrenia.


Subject(s)
Antipsychotic Agents/administration & dosage , Catechols/administration & dosage , Nitriles/administration & dosage , Schizophrenia/diagnosis , Schizophrenia/drug therapy , Schizophrenic Psychology , Adolescent , Adult , Catechol O-Methyltransferase/metabolism , Catechol O-Methyltransferase Inhibitors , Double-Blind Method , Drug Therapy, Combination , Enzyme Inhibitors/administration & dosage , Female , Humans , Male , Middle Aged , Schizophrenia/enzymology , Treatment Outcome , Young Adult
20.
J Biol Chem ; 289(4): 2318-30, 2014 Jan 24.
Article in English | MEDLINE | ID: mdl-24302717

ABSTRACT

There is compelling evidence that aminoglycoside (AG) antibiotics can induce the mammalian ribosome to suppress disease-causing nonsense mutations and partially restore the expression of functional proteins. However, prolonged AG treatment can cause detrimental side effects in patients, including most prominently, ototoxicity. Recent mechanistic discussions have considered the relative contributions of mitochondrial and cytoplasmic protein synthesis inhibition to AG-induced ototoxicity. We show that AGs inhibit mitochondrial protein synthesis in mammalian cells and perturb cell respiration, leading to a time- and dose-dependent increase in superoxide overproduction and accumulation of free ferrous iron in mitochondria caused by oxidative damage of mitochondrial aconitase, ultimately leading to cell apoptosis via the Fenton reaction. These deleterious effects increase with the increased potency of AG to inhibit the mitochondrial rather than cytoplasmic protein synthesis, which in turn correlates with their ototoxic potential in both murine cochlear explants and the guinea pig in vivo. The deleterious effects of AGs were alleviated in synthetic derivatives specially designed for the treatment of genetic diseases caused by nonsense mutations and possessing low affinity toward mitochondrial ribosomes. This work highlights the benefit of a mechanism-based drug redesign strategy that can maximize the translational value of "readthrough therapy" while mitigating drug-induced side effects. This approach holds promise for patients suffering from genetic diseases caused by nonsense mutations.


Subject(s)
Aminoglycosides/pharmacology , Cytoplasm/metabolism , Mitochondria/metabolism , Protein Synthesis Inhibitors/pharmacology , Ribosomes/metabolism , Aminoglycosides/adverse effects , Animals , Apoptosis/drug effects , Cochlea/metabolism , Dose-Response Relationship, Drug , Genetic Diseases, Inborn/drug therapy , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Guinea Pigs , HeLa Cells , Humans , Mice , Mitochondrial Proteins/biosynthesis , Oxygen Consumption/drug effects , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/adverse effects , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...