Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 7376, 2024 03 28.
Article in English | MEDLINE | ID: mdl-38548767

ABSTRACT

CDKL5 deficiency disorder (CDD) is a neurodevelopmental condition characterized by global developmental delay, early-onset seizures, intellectual disability, visual and motor impairments. Unlike Rett Syndrome (RTT), CDD lacks a clear regression period. Patients with CDD frequently encounter gastrointestinal (GI) disturbances and exhibit signs of subclinical immune dysregulation. However, the underlying causes of these conditions remain elusive. Emerging studies indicate a potential connection between neurological disorders and gut microbiota, an area completely unexplored in CDD. We conducted a pioneering study, analyzing fecal microbiota composition in individuals with CDD (n = 17) and their healthy relatives (n = 17). Notably, differences in intestinal bacterial diversity and composition were identified in CDD patients. In particular, at genus level, CDD microbial communities were characterized by an increase in the relative abundance of Clostridium_AQ, Eggerthella, Streptococcus, and Erysipelatoclostridium, and by a decrease in Eubacterium, Dorea, Odoribacter, Intestinomonas, and Gemmiger, pointing toward a dysbiotic profile. We further investigated microbiota changes based on the severity of GI issues, seizure frequency, sleep disorders, food intake type, impairment in neuro-behavioral features and ambulation capacity. Enrichment in Lachnoclostridium and Enterobacteriaceae was observed in the microbiota of patients with more severe GI symptoms, while Clostridiaceae, Peptostreptococcaceae, Coriobacteriaceae, Erysipelotrichaceae, Christensenellaceae, and Ruminococcaceae were enriched in patients experiencing daily epileptic seizures. Our findings suggest a potential connection between CDD, microbiota and symptom severity. This study marks the first exploration of the gut-microbiota-brain axis in subjects with CDD. It adds to the growing body of research emphasizing the role of the gut microbiota in neurodevelopmental disorders and opens doors to potential interventions that target intestinal microbes with the aim of improving the lives of patients with CDD.


Subject(s)
Epileptic Syndromes , Gastrointestinal Microbiome , Rett Syndrome , Spasms, Infantile , Humans , Gastrointestinal Microbiome/physiology , Rett Syndrome/genetics , Seizures , Protein Serine-Threonine Kinases
2.
Cell Rep ; 42(7): 112788, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37436896

ABSTRACT

Perineuronal nets (PNNs) surround specific neurons in the brain and are involved in various forms of plasticity and clinical conditions. However, our understanding of the PNN role in these phenomena is limited by the lack of highly quantitative maps of PNN distribution and association with specific cell types. Here, we present a comprehensive atlas of Wisteria floribunda agglutinin (WFA)-positive PNNs and colocalization with parvalbumin (PV) cells for over 600 regions of the adult mouse brain. Data analysis shows that PV expression is a good predictor of PNN aggregation. In the cortex, PNNs are dramatically enriched in layer 4 of all primary sensory areas in correlation with thalamocortical input density, and their distribution mirrors intracortical connectivity patterns. Gene expression analysis identifies many PNN-correlated genes. Strikingly, PNN-anticorrelated transcripts are enriched in synaptic plasticity genes, generalizing PNNs' role as circuit stability factors.


Subject(s)
Extracellular Matrix , Parvalbumins , Animals , Mice , Parvalbumins/metabolism , Mice, Inbred C57BL , Extracellular Matrix/metabolism , Neurons/metabolism , Cerebral Cortex/metabolism
4.
Neuropharmacology ; 231: 109491, 2023 06 15.
Article in English | MEDLINE | ID: mdl-36924923

ABSTRACT

Neuroplasticity refers to the ability of brain circuits to reorganize and change the properties of the network, resulting in alterations in brain function and behavior. It is traditionally believed that neuroplasticity is influenced by external stimuli, learning, and experience. Intriguingly, there is new evidence suggesting that endogenous signals from the body's periphery may play a role. The gut microbiota, a diverse community of microorganisms living in harmony with their host, may be able to influence plasticity through its modulation of the gut-brain axis. Interestingly, the maturation of the gut microbiota coincides with critical periods of neurodevelopment, during which neural circuits are highly plastic and potentially vulnerable. As such, dysbiosis (an imbalance in the gut microbiota composition) during early life may contribute to the disruption of normal developmental trajectories, leading to neurodevelopmental disorders. This review aims to examine the ways in which the gut microbiota can affect neuroplasticity. It will also discuss recent research linking gastrointestinal issues and bacterial dysbiosis to various neurodevelopmental disorders and their potential impact on neurological outcomes. This article is part of the Special Issue on "Microbiome & the Brain: Mechanisms & Maladies".


Subject(s)
Gastrointestinal Microbiome , Neurodevelopmental Disorders , Humans , Dysbiosis , Brain , Neuronal Plasticity
5.
Cell Mol Life Sci ; 80(1): 28, 2023 Jan 06.
Article in English | MEDLINE | ID: mdl-36607453

ABSTRACT

Little is known about the impact of metabolic stimuli on brain tissue at a molecular level. The ketone body beta-hydroxybutyrate (BHB) can be a signaling molecule regulating gene transcription. Thus, we assessed lysine beta-hydroxybutyrylation (K-bhb) levels in proteins extracted from the cerebral cortex of mice undergoing a ketogenic metabolic challenge (48 h fasting). We found that fasting enhanced K-bhb in a variety of proteins including histone H3. ChIP-seq experiments showed that K9 beta-hydroxybutyrylation of H3 (H3K9-bhb) was significantly enriched by fasting on more than 8000 DNA loci. Transcriptomic analysis showed that H3K9-bhb on enhancers and promoters correlated with active gene expression. One of the most enriched functional annotations both at the epigenetic and transcriptional level was "circadian rhythms''. Indeed, we found that the diurnal oscillation of specific transcripts was modulated by fasting at distinct zeitgeber times both in the cortex and suprachiasmatic nucleus. Moreover, specific changes in locomotor activity daily features were observed during re-feeding after 48-h fasting. Thus, our results suggest that fasting remarkably impinges on the cerebral cortex transcriptional and epigenetic landscape, and BHB acts as a powerful epigenetic molecule in the brain through direct and specific histone marks remodeling in neural tissue cells.


Subject(s)
Histones , Ketone Bodies , Mice , Animals , Histones/metabolism , 3-Hydroxybutyric Acid/metabolism , Ketone Bodies/metabolism , Brain/metabolism , Gene Expression
6.
Int J Mol Sci ; 23(16)2022 Aug 10.
Article in English | MEDLINE | ID: mdl-36012156

ABSTRACT

Among Histone post-translational modifications (PTMs), lysine acetylation plays a pivotal role in the epigenetic regulation of gene expression, mediated by chromatin modifying enzymes. Due to their activity in physiology and pathology, several chemical compounds have been developed to inhibit the function of these proteins. However, the pleiotropy of these classes of proteins represents a weakness of epigenetic drugs. Ideally, a new generation of epigenetic drugs should target with molecular precision individual acetylated lysines on the target protein. We exploit a PTM-directed interference, based on an intrabody (scFv-58F) that selectively binds acetylated lysine 9 of histone H3 (H3K9ac), to test the hypothesis that targeting H3K9ac yields more specific effects than inhibiting the corresponding HAT enzyme that installs that PTM. In yeast scFv-58F modulates, gene expression in a more specific way, compared to two well-established HAT inhibitors. This PTM-specific interference modulated expression of genes involved in ribosome biogenesis and function. In mammalian cells, the scFv-58F induces exclusive changes in the H3K9ac-dependent expression of specific genes. These results suggest the H3K9ac-specific intrabody as the founder of a new class of molecules to directly target histone PTMs, inverting the paradigm from inhibiting the writer enzyme to acting on the PTM.


Subject(s)
Histones , Lysine , Acetylation , Animals , Epigenesis, Genetic , Gene Expression , Histone Acetyltransferases/metabolism , Histones/metabolism , Lysine/metabolism , Mammals/metabolism , Protein Processing, Post-Translational
7.
Nutrients ; 14(4)2022 Feb 13.
Article in English | MEDLINE | ID: mdl-35215432

ABSTRACT

The consumption of a high-fat, low-carbohydrate diet (ketogenic diet) has diverse effects on health and is expected to have therapeutic value in neurological disorders, metabolic syndrome, and cancer. Recent studies have shown that a ketogenic diet not only pronouncedly shifts the cellular metabolism to pseudo-starvation, but also exerts a variety of physiological functions on various organs through metabolites that act as energy substrates, signaling molecules, and epigenetic modifiers. In this review, we highlight the latest findings on the molecular mechanisms of a ketogenic diet and speculate on the significance of these functions in the context of the epigenome and microbiome. Unraveling the molecular basis of the bioactive effects of a ketogenic diet should provide solid evidence for its clinical application in a variety of diseases including cancer.


Subject(s)
Diet, Ketogenic , Microbiota , Nervous System Diseases , Diet, Carbohydrate-Restricted , Diet, High-Fat , Humans
8.
Cell Rep ; 38(2): 110212, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35021093

ABSTRACT

Exposing animals to an enriched environment (EE) has dramatic effects on brain structure, function, and plasticity. The poorly known "EE-derived signals'' mediating the EE effects are thought to be generated within the central nervous system. Here, we shift the focus to the body periphery, revealing that gut microbiota signals are crucial for EE-driven plasticity. Developmental analysis reveals striking differences in intestinal bacteria composition between EE and standard rearing (ST) mice, as well as enhanced levels of short-chain fatty acids (SCFA) in EE mice. Depleting the microbiota of EE mice with antibiotics strongly decreases SCFA and prevents activation of adult ocular dominance plasticity, spine dynamics, and microglia rearrangement. SCFA treatment in ST mice mimics EE induction of ocular dominance plasticity and microglial remodeling. Remarkably, transferring the microbiota of EE mice to ST recipients activates adult ocular dominance plasticity. Thus, experience-dependent changes in gut microbiota regulate brain plasticity.


Subject(s)
Gastrointestinal Microbiome/physiology , Neuronal Plasticity/physiology , Visual Cortex/metabolism , Animals , Brain/physiology , Dominance, Ocular/physiology , Environment , Fatty Acids, Volatile/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Visual Cortex/microbiology
9.
Proc Natl Acad Sci U S A ; 117(47): 29904-29913, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33172990

ABSTRACT

Food is a powerful entrainment cue for circadian clocks in peripheral tissues, and changes in the composition of nutrients have been demonstrated to metabolically reprogram peripheral clocks. However, how food challenges may influence circadian metabolism of the master clock in the suprachiasmatic nucleus (SCN) or in other brain areas is poorly understood. Using high-throughput metabolomics, we studied the circadian metabolome profiles of the SCN and medial prefrontal cortex (mPFC) in lean mice compared with mice challenged with a high-fat diet (HFD). Both the mPFC and the SCN displayed a robust cyclic metabolism, with a strikingly high sensitivity to HFD perturbation in an area-specific manner. The phase and amplitude of oscillations were drastically different between the SCN and mPFC, and the metabolic pathways impacted by HFD were remarkably region-dependent. Furthermore, HFD induced a significant increase in the number of cycling metabolites exclusively in the SCN, revealing an unsuspected susceptibility of the master clock to food stress.


Subject(s)
Circadian Clocks/physiology , Diet, High-Fat/adverse effects , Metabolome/physiology , Prefrontal Cortex/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Male , Metabolomics , Mice , Models, Animal , Photoperiod
10.
EMBO Rep ; 21(11): e50431, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33026181

ABSTRACT

Visual cortical circuits show profound plasticity during early life and are later stabilized by molecular "brakes" limiting excessive rewiring beyond a critical period. The mechanisms coordinating the expression of these factors during the transition from development to adulthood remain unknown. We found that miR-29a expression in the visual cortex dramatically increases with age, but it is not experience-dependent. Precocious high levels of miR-29a blocked ocular dominance plasticity and caused an early appearance of perineuronal nets. Conversely, inhibition of miR-29a in adult mice using LNA antagomirs activated ocular dominance plasticity, reduced perineuronal nets, and restored their juvenile chemical composition. Activated adult plasticity had the typical functional and proteomic signature of critical period plasticity. Transcriptomic and proteomic studies indicated that miR-29a manipulation regulates the expression of plasticity brakes in specific cortical circuits. These data indicate that miR-29a is a regulator of the plasticity brakes promoting age-dependent stabilization of visual cortical connections.


Subject(s)
MicroRNAs , Visual Cortex , Animals , Dominance, Ocular/genetics , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Neuronal Plasticity/genetics , Proteomics
11.
Nat Commun ; 11(1): 4448, 2020 09 07.
Article in English | MEDLINE | ID: mdl-32895370

ABSTRACT

Substance abuse disorders are linked to alteration of circadian rhythms, although the molecular and neuronal pathways implicated have not been fully elucidated. Addictive drugs, such as cocaine, induce a rapid increase of dopamine levels in the brain. Here, we show that acute administration of cocaine triggers reprogramming in circadian gene expression in the striatum, an area involved in psychomotor and rewarding effects of drugs. This process involves the activation of peroxisome protein activator receptor gamma (PPARγ), a nuclear receptor involved in inflammatory responses. PPARγ reprogramming is altered in mice with cell-specific ablation of the dopamine D2 receptor (D2R) in the striatal medium spiny neurons (MSNs) (iMSN-D2RKO). Administration of a specific PPARγ agonist in iMSN-D2RKO mice elicits substantial rescue of cocaine-dependent control of circadian genes. These findings have potential implications for development of strategies to treat substance abuse disorders.


Subject(s)
Circadian Clocks/drug effects , Cocaine-Related Disorders/physiopathology , Cocaine/adverse effects , Nucleus Accumbens/drug effects , PPAR gamma/metabolism , Receptors, Dopamine D2/metabolism , Administration, Oral , Animals , Circadian Clocks/physiology , Cocaine/administration & dosage , Cocaine-Related Disorders/drug therapy , Dopamine/metabolism , Injections, Intraperitoneal , Locomotion/physiology , Male , Mice , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Nucleus Accumbens/physiopathology , PPAR gamma/agonists , Pioglitazone/administration & dosage , Receptors, Dopamine D2/genetics , Reward , Signal Transduction
12.
Genes (Basel) ; 11(7)2020 07 03.
Article in English | MEDLINE | ID: mdl-32635190

ABSTRACT

Gene expression in the brain is dramatically regulated by a variety of stimuli. While the role of neural activity has been extensively studied, less is known about the effects of metabolism and nutrition on transcriptional control mechanisms in the brain. Extracellular signals are integrated at the chromatin level through dynamic modifications of epigenetic marks, which in turn fine-tune gene transcription. In the last twenty years, it has become clear that epigenetics plays a crucial role in modulating central nervous system functions and finally behavior. Here, we will focus on the effect of metabolic signals in shaping brain DNA methylation, both during development and adulthood. We will provide an overview of maternal nutrition effects on brain methylation and behavior in offspring. In addition, the impact of different diet challenges on cytosine methylation dynamics in the adult brain will be discussed. Finally, the possible role played by the metabolic status in modulating DNA hydroxymethylation, which is particularly abundant in neural tissue, will be considered.


Subject(s)
Brain/metabolism , DNA Methylation , Epigenesis, Genetic , Prenatal Exposure Delayed Effects/genetics , Prenatal Nutritional Physiological Phenomena , Animals , Brain/embryology , Brain/physiology , Female , Pregnancy
13.
Article in English | MEDLINE | ID: mdl-32039048

ABSTRACT

Advances in high-throughput sequencing technologies in the past decade has led to a tremendous growth in knowledge about the role played by microorganisms on our body health. Trillions of microbes live in close symbiosis with their host, and have impacts on various aspects of host physiology as well as predisposition to disease. This is a consequence of the direct interaction between host cells and microbes or their signaling molecules, such as metabolites, which can reach and exert their effects in distal tissues. Among the essential factors modulating the human body's ecosystem of symbionts, the circadian clock might be one of the key regulators. The endogenous clock is a highly conserved timekeeper able to align organismal physiology to the daily cycle, thus maximizing survival and fitness. Circadian rhythms coordinate whole-body biological processes synchronizing cellular biochemical reactions, tissue function and finally controlling systemic homeostasis. Intriguingly, growing body of evidence has demonstrated that the host circadian cycle governs the structure of the gut microbiota community and its diurnal rhythmicity, whereas the microbes contribute to maintenance of clock function. In this review, we will give an overview of the multisystem aspects of microbiome-host interactions in the context of circadian rhythmicity. In particular, the effect of the interaction clock-microbial communities on immune system function and metabolic homeostasis will be discussed. Finally, the possible implication of daily rhythm on the gut-microbiome-brain axis will be analyzed, focusing on the reciprocal effects of clock disruption and microbiota alterations on brain function and behavior.


Subject(s)
Circadian Clocks , Gastrointestinal Microbiome , Host Microbial Interactions , Symbiosis , Animals , Humans
14.
Cell ; 174(6): 1571-1585.e11, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30193114

ABSTRACT

Metabolic diseases are often characterized by circadian misalignment in different tissues, yet how altered coordination and communication among tissue clocks relate to specific pathogenic mechanisms remains largely unknown. Applying an integrated systems biology approach, we performed 24-hr metabolomics profiling of eight mouse tissues simultaneously. We present a temporal and spatial atlas of circadian metabolism in the context of systemic energy balance and under chronic nutrient stress (high-fat diet [HFD]). Comparative analysis reveals how the repertoires of tissue metabolism are linked and gated to specific temporal windows and how this highly specialized communication and coherence among tissue clocks is rewired by nutrient challenge. Overall, we illustrate how dynamic metabolic relationships can be reconstructed across time and space and how integration of circadian metabolomics data from multiple tissues can improve our understanding of health and disease.


Subject(s)
Circadian Clocks/physiology , Metabolome , Animals , Diet, High-Fat , Energy Metabolism , Liver/metabolism , Male , Metabolic Networks and Pathways , Metabolomics , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Prefrontal Cortex/metabolism , Suprachiasmatic Nucleus/metabolism , Uncoupling Protein 1/metabolism
15.
Article in English | MEDLINE | ID: mdl-29038112

ABSTRACT

Circadian rhythms influence virtually all life forms on our planet, a notion that opens the question on how the circadian cycles of individual organisms may interplay with each other. In mammals, a potentially dangerous environmental stress is represented by encounters with infectious agents. Microbial attack is a major risk for organismal homeostasis and therefore needs to be efficiently counteracted by mechanisms implemented by the host immune system. Accumulating evidence shows that the immune system may anticipate an emerging pathogenic exposure through an enhanced inflammatory state. Notably, the circadian clock orchestrates these anticipatory responses to fluctuating conditions in the external world. In this article, we review the current knowledge about the relationship between the circadian clock and pathogenic infections. We discuss the role of the circadian clock against infection and specific pathogens, the core clock proteins involved in the defense mechanisms, and the specific tissue or cell type in which they function to counteract the infection. Finally, circadian oscillations in the gut microbiome composition and its possible role in protecting against foodborne pathogen colonization are presented.


Subject(s)
Circadian Clocks/physiology , Gastrointestinal Microbiome/physiology , Animals , Circadian Rhythm , Food Microbiology , Inflammation/immunology , Inflammation/microbiology , Inflammation/virology
16.
Cell Metab ; 26(3): 523-538.e5, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28877456

ABSTRACT

The circadian clock orchestrates rhythms in physiology and behavior, allowing organismal adaptation to daily environmental changes. While food intake profoundly influences diurnal rhythms in the liver, how nutritional challenges are differentially interpreted by distinct tissue-specific clocks remains poorly explored. Ketogenic diet (KD) is considered to have metabolic and therapeutic value, though its impact on circadian homeostasis is virtually unknown. We show that KD has profound and differential effects on liver and intestine clocks. Specifically, the amplitude of clock-controlled genes and BMAL1 chromatin recruitment are drastically altered by KD in the liver, but not in the intestine. KD induces nuclear accumulation of PPARα in both tissues but with different circadian phase. Also, gut and liver clocks respond differently to carbohydrate supplementation to KD. Importantly, KD induces serum and intestinal ß-hydroxyl-butyrate levels to robustly oscillate in a circadian manner, an event coupled to tissue-specific cyclic histone deacetylase (HDAC) activity and histone acetylation.


Subject(s)
Circadian Clocks , Circadian Rhythm , Diet, Ketogenic , Gastrointestinal Tract/physiology , Liver/physiology , 3-Hydroxybutyric Acid/blood , 3-Hydroxybutyric Acid/metabolism , ARNTL Transcription Factors/metabolism , Acetylation/drug effects , Animals , Chromatin/metabolism , Circadian Clocks/drug effects , Circadian Clocks/genetics , Circadian Rhythm/drug effects , Circadian Rhythm/genetics , Epigenesis, Genetic/drug effects , Fructose/pharmacology , Gene Knockdown Techniques , Histones/metabolism , Liver/drug effects , Metabolome/genetics , Mice , Models, Biological , Organ Specificity/drug effects , Organ Specificity/genetics , PPAR alpha/metabolism , Promoter Regions, Genetic/genetics , Respiration/drug effects , Sucrose/pharmacology , Transcription, Genetic/drug effects , Transcriptome/genetics
17.
Cell Host Microbe ; 22(2): 185-192, 2017 Aug 09.
Article in English | MEDLINE | ID: mdl-28799904

ABSTRACT

Microbial infection poses a threat to organismal homeostasis and therefore must be efficiently counteracted by host defense mechanisms. It has been recently demonstrated that the immune system may anticipate an emerging pathogenic exposure through a heightened inflammatory state. Such anticipatory responses to fluctuating environmental conditions are typically orchestrated by the circadian clock, an intrinsic time-keeping system that adapts tissue physiology to diurnal variations in external influences. Here, we review current knowledge about the interplay between the circadian clock and antimicrobial responses. We summarize the molecular strategies employed by the circadian system against specific pathogens, the core-clock proteins as well as cells in which they are expressed that mediate host defense, and the consequences of circadian variations on immune function. Furthermore, we highlight the possible implications of such circadian gating in immune reactions against pathogenic infections for the chronopharmacology of antibacterial and antiviral therapies.


Subject(s)
Anti-Infective Agents/immunology , Circadian Clocks/physiology , Circadian Rhythm/physiology , Antiviral Agents/immunology , Homeostasis , Humans , Immune System/physiology , Immunity , Microbiota
18.
Nat Commun ; 8: 15488, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28534484

ABSTRACT

MicroRNAs (miRNAs) are known to mediate post-transcriptional gene regulation, but their role in postnatal brain development is still poorly explored. We show that the expression of many miRNAs is dramatically regulated during functional maturation of the mouse visual cortex with miR-132/212 family being one of the top upregulated miRNAs. Age-downregulated transcripts are significantly enriched in miR-132/miR-212 putative targets and in genes upregulated in miR-132/212 null mice. At a functional level, miR-132/212 deletion affects development of receptive fields of cortical neurons determining a specific impairment of binocular matching of orientation preference, but leaving orientation and direction selectivity unaltered. This deficit is associated with reduced depth perception in the visual cliff test. Deletion of miR-132/212 from forebrain excitatory neurons replicates the binocular matching deficits. Thus, miR-132/212 family shapes the age-dependent transcriptome of the visual cortex during a specific developmental window resulting in maturation of binocular cortical cells and depth perception.


Subject(s)
Depth Perception , MicroRNAs/physiology , Orientation , Visual Perception , Animals , Electrophysiology , Female , Gene Deletion , Genotype , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Neuronal Plasticity , Neurons/physiology , Sequence Analysis, RNA , Transcriptome , Up-Regulation , Vision, Binocular , Visual Cortex
19.
Front Cell Neurosci ; 11: 25, 2017.
Article in English | MEDLINE | ID: mdl-28223922

ABSTRACT

During childhood, our brain is exposed to a variety of environmental inputs that can sculpt synaptic connections and neuronal circuits, with subsequent influence on behavior and learning processes. Critical periods of neurodevelopment are windows of opportunity in which the neuronal circuits are extremely plastic and can be easily subjected to remodeling in response to experience. However, the brain is also more susceptible to aberrant stimuli that might lead to altered developmental trajectories. Intriguingly, postnatal brain development is paralleled by the maturation of the gut microbiota: the ecosystem of symbionts populating our gastro-intestinal tract. Recent discoveries have started to unveil an unexpected link between the gut microbiome and neurophysiological processes. Indeed, the commensal bacteria seem to be able to influence host behavioral outcome and neurochemistry through mechanisms which remain poorly understood. Remarkably, the efficacy of the gut flora action appears to be dependent on the timing during postnatal life at which the host gut microbes' signals reaches the brain, suggesting the fascinating possibility of critical periods for this microbiota-driven shaping of host neuronal functions and behavior. Therefore, to understand the importance of the intestinal ecosystem's impact on neuronal circuits functions and plasticity during development and the discovery of the involved molecular mechanisms, will pave the way to identify new and, hopefully, powerful microbiota-based therapeutic interventions for the treatment of neurodevelopmental and psychiatric diseases.

20.
EMBO Rep ; 17(9): 1292-303, 2016 09.
Article in English | MEDLINE | ID: mdl-27418314

ABSTRACT

The liver circadian clock is reprogrammed by nutritional challenge through the rewiring of specific transcriptional pathways. As the gut microbiota is tightly connected to host metabolism, whose coordination is governed by the circadian clock, we explored whether gut microbes influence circadian homeostasis and how they distally control the peripheral clock in the liver. Using fecal transplant procedures we reveal that, in response to high-fat diet, the gut microbiota drives PPARγ-mediated activation of newly oscillatory transcriptional programs in the liver. Moreover, antibiotics treatment prevents PPARγ-driven transcription in the liver, underscoring the essential role of gut microbes in clock reprogramming and hepatic circadian homeostasis. Thus, a specific molecular signature characterizes the influence of the gut microbiome in the liver, leading to the transcriptional rewiring of hepatic metabolism.


Subject(s)
Circadian Clocks , Diet, High-Fat , Gastrointestinal Microbiome , Liver/metabolism , PPAR gamma/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Blood Glucose , Circadian Clocks/drug effects , Circadian Clocks/genetics , Circadian Rhythm , Cluster Analysis , Energy Metabolism/genetics , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/drug effects , Gene Expression Profiling , Humans , Male , Mice , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...