Gut
gut
From heterogeneous wiring to degenerative function in motion-detection circuits
Gut/Body interactions in health and disease
The adult intestine is a major barrier epithelium and coordinator of multi-organ functions. Stem cells constantly repair the intestinal epithelium by adjusting their proliferation and differentiation to tissue intrinsic as well as micro- and macro-environmental signals. How these signals integrate to control intestinal and whole-body homeostasis is largely unknown. Addressing this gap in knowledge is central to an improved understanding of intestinal pathophysiology and its systemic consequences. Combining Drosophila and mammalian model systems my laboratory has discovered fundamental mechanisms driving intestinal regeneration and tumourigenesis and outlined complex inter-organ signaling regulating health and disease. During my talk, I will discuss inter-related areas of research from my lab, including:1- Interactions between the intestine and its microenvironment influencing intestinal regeneration and tumourigenesis. 2- Long-range signals from the intestine impacting whole-body in health and disease.
From the guts to the brain through adaptive immunity in the prevention of Alzheimer’ disease
Dr. Pasinetti is the Saunders Family Chair and Professor of Neurology at Icahn School of medicine at Mount Sinai, New York. His studies allowed him to develop novel therapeutic approaches through investigation of preventable risk factors including mood disorders in the promotion of resilience against neurodegenerative disorder. In his presentation Dr. Pasinetti will discuss novel concepts about the gut-brain axis in mechanisms associated to peripheral adaptive immunity as therapeutic targets to mitigate the onset and the progression of Alzheimer’s disease and other form of dementia.
Identification of dendritic cell-T cell interactions driving immune responses to food
A microbiome-dependent gut-brain pathway regulates motivation for exercise
PIEZO2 in somatosensory neurons coordinates gastrointestinal transit
The transit of food through the gastrointestinal tract is critical for nutrient absorption and survival, and the gastrointestinal tract has the ability to initiate motility reflexes triggered by luminal distention. This complex function depends on the crosstalk between extrinsic and intrinsic neuronal innervation within the intestine, as well as local specialized enteroendocrine cells. However, the molecular mechanisms and the subset of sensory neurons underlying the initiation and regulation of intestinal motility remain largely unknown. Here, we show that humans lacking PIEZO2 exhibit impaired bowel sensation and motility. Piezo2 in mouse dorsal root but not nodose ganglia is required to sense gut content, and this activity slows down food transit rates in the stomach, small intestine, and colon. Indeed, Piezo2 is directly required to detect colon distension in vivo. Our study unveils the mechanosensory mechanisms that regulate the transit of luminal contents throughout the gut, which is a critical process to ensure proper digestion, nutrient absorption, and waste removal. These findings set the foundation of future work to identify the highly regulated interactions between sensory neurons, enteric neurons and non- neuronal cells that control gastrointestinal motility.
Immune regulation by fungal strain diversity in inflammatory bowel disease
Gut-brain circuits for fat preference
Gut-to-brain communication of nutritional information prioritizes courtship over feeding
The person-to-person transmission landscape of the gut and oral microbiomes
Nociceptor neurons direct goblet cells via a CGRP-RAMP1 axis to drive mucus production and gut barrier protection
Beta oscillations in the basal ganglia: Past, Present and Future; Oscillatory signatures of motor symptoms across movement disorders
On Wednesday, January 25th, at noon ET / 6PM CET, we will host Roxanne Lofredi and Hagai Bergman. Roxanne Lofredi, MD, is a research fellow in the Movement Disorders and Neuromodulation Unit at Charité Universitätsmedizin Berlin. Hagai Bergman, MD, PhD, is a Professor of Physiology in the Edmond and Lily Safra Center for Brain Research and Faculty of Medicine at the Hebrew University of Jerusalem, and is Simone and Bernard Guttman Chair in Brain Research. Beside his scientific presentation on “Beta oscillations in the basal ganglia: Past, Present and Future”, he will also give us a glimpse at the “Person behind the science”. The talks will be followed by a shared discussion. You can register via talks.stimulatingbrains.org to receive the (free) Zoom link!
How can we treat visceral pain?
Chronic pain is a leading cause of morbidity, common to patients with gastrointestinal diseases such as irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD). Most pain killers are largely ineffective against this type of pain or restricted for use in these patients due to gut related complications and risk of addition. A significant unmet clinical need therefore exists to develop novel non-opioid based visceral analgesics.
Gut food cravings? How gut signals control appetite and metabolism
Gut-derived signals regulate metabolism, appetite, and behaviors important for mental health. We have performed a large-scale multidimensional screen to identify gut hormones and nutrient-sensing mechanisms in the intestine that regulate metabolism and behavior in the fruit fly Drosophila. We identified several gut hormones that affect fecundity, stress responses, metabolism, feeding, and sleep behaviors, many of which seem to act sex-specifically. We show that in response to nutrient intake, the enteroendocrine cells (EECs) of the adult Drosophila midgut release hormones that act via inter-organ relays to coordinate metabolism and feeding decisions. These findings suggest that crosstalk between the gut and other tissues regulates food choice according to metabolic needs, providing insight into how that intestine processes nutritional inputs and into the gut-derived signals that relay information regulating nutrient-specific hungers to maintain metabolic homeostasis.
Gut Feelings: The Microbiome as a Key Regulator of Brain & Behaviour Across the Lifespan
Inter-tissue signals modify food-seeking behavior in C. elegans
Animals modify their behavioral outputs in response to changes in external and internal environments. We use the nematode, C. elegans to probe the pathways linking changes in internal states like hunger with behavior. We find that acute food deprivation alters the localization of two transcription factors, likely releasing an insulin-like peptide from the intestine, which in turn modifies chemosensory neurons and alters behavior. These results present a model for how inter-tissue signals to generate flexible behaviors via gut-brain signaling.
Western diet consumption and memory impairment: what, when, and how?
Habitual consumption of a “Western diet”, containing higher than recommended levels of simple sugars and saturated fatty acids, is associated with cognitive impairments in humans and in various experimental animal models. Emerging findings reveal that the specific mnemonic processes that are disrupted by Western diet consumption are those that rely on the hippocampus, a brain region classically linked with memory control and more recently with the higher-order control of food intake. Our laboratory has established rat models in which excessive consumption of different components of a Western diet during the juvenile and adolescent periods of development yields long-term impairments in hippocampal-dependent memory function without concomitant increases in total caloric intake, body weight, or adiposity. Our ongoing work is investigating alterations in the gut microbiome as a potential underlying neurobiological mechanism linking early life unhealthy dietary factors to adverse neurocognitive outcomes.
Neural Circuit Dysfunction along the Gut/Brain Axis in zebrafish models of Autism Spectrum Disorder
Plasticity in gut microbe-host interactions
Plasticity in gut microbe-host interactions
Role of the gut microbiota in the development of alcohol use disorder
The gut microbiota is composed of a very large number of bacteria, viruses, fungi and yeasts that play an important role in the body, through the production of a series of metabolites (including neurotransmitters), and through an essential role in the barrier function of the gut and the regulation of immunity and stress response. In this lecture I will present, based mainly on human studies but also on preclinical studies, the evidence for a role of the gut microbiota in the development of alcohol use disorder. I will show the first results of trials to test the effects of nutritional approaches to address these deficits.
Adapt or Die: Transgenerational Inheritance of Pathogen Avoidance (or, How getting food poisoning might save your species)
Caenorhabditis elegans must distinguish pathogens from nutritious food sources among the many bacteria to which it is exposed in its environment1. Here we show that a single exposure to purified small RNAs isolated from pathogenic Pseudomonas aeruginosa (PA14) is sufficient to induce pathogen avoidance in the treated worms and in four subsequent generations of progeny. The RNA interference (RNAi) and PIWI-interacting RNA (piRNA) pathways, the germline and the ASI neuron are all required for avoidance behaviour induced by bacterial small RNAs, and for the transgenerational inheritance of this behaviour. A single P. aeruginosa non-coding RNA, P11, is both necessary and sufficient to convey learned avoidance of PA14, and its C. elegans target, maco-1, is required for avoidance. Our results suggest that this non-coding-RNA-dependent mechanism evolved to survey the microbial environment of the worm, use this information to make appropriate behavioural decisions and pass this information on to its progeny.
Gut-brain signaling as a driver of behavior and gene expression in a mouse model for autism spectrum disorder
In vitro bioelectronic models of the gut-brain axis
The human gut microbiome has emerged as a key player in the bidirectional communication of the gut-brain axis, affecting various aspects of homeostasis and pathophysiology. Until recently, the majority of studies that seek to explore the mechanisms underlying the microbiome-gut-brain axis cross-talk relied almost exclusively on animal models, and particularly gnotobiotic mice. Despite the great progress made with these models, various limitations, including ethical considerations and interspecies differences that limit the translatability of data to human systems, pushed researchers to seek for alternatives. Over the past decades, the field of in vitro modelling of tissues has experienced tremendous growth, thanks to advances in 3D cell biology, materials, science and bioengineering, pushing further the borders of our ability to more faithfully emulate the in vivo situation. Organ-on-chip technology and bioengineered tissues have emerged as highly promising alternatives to animal models for a wide range of applications. In this talk I’ll discuss our progress towards generating a complete platform of the human microbiota-gut-brain axis with integrated monitoring and sensing capabilities. Bringing together principles of materials science, tissue engineering, 3D cell biology and bioelectronics, we are building advanced models of the GI and the BBB /NVU, with real-time and label-free monitoring units adapted in the model architecture, towards a robust and more physiologically relevant human in vitro model, aiming to i) elucidate the role of microbiota in the gut-brain axis communication, ii) to study how diet and impaired microbiota profiles affect various (patho-)physiologies, and iii) to test personalised medicine approaches for disease modelling and drug testing.
How much gut needs the brain ? Gut microbiota-immune crosstalk in neuroinflammation
Gestational exposure to environmental toxins, infections, and stressors are epidemiologically linked to neurodevelopmental disorders
Gestational exposure to environmental toxins, infections, and stressors are epidemiologically linked to neurodevelopmental disorders with strong male-bias, such as autism spectrum disorder. We modeled some of these prenatal risk factors in mice, by co-exposing pregnant dams to an environmental pollutant and limited-resource stress, which robustly dysregulated the maternal immune system. Male but not female offspring displayed long-lasting behavioral abnormalities and alterations in the activity of brain networks encoding social interactions, along with disruptions of gut structure and microbiome composition. Cellularly, prenatal stressors impaired microglial synaptic pruning in males during early postnatal development. Precise inhibition of microglial phagocytosis during the same critical period mimicked the impact of prenatal stressors on the male-specific social deficits. Conversely, modifying the gut microbiome rescued the social and cellular deficits, indicating that environmental stressors alter neural circuit formation in males via impairing microglia function during development, perhaps via a gut-brain disruption.
Disinhibitory and neuromodulatory regulation of hippocampal synaptic plasticity
The CA1 pyramidal neurons are embedded in an intricate local circuitry that contains a variety of interneurons. The roles these interneurons play in the regulation of the excitatory synaptic plasticity remains largely understudied. Recent experiments showed that repeated cholinergic activation of 𝛼7 nACh receptors expressed in oriens-lacunosum-moleculare (OLM𝛼2) interneurons could induce LTP in SC-CA1 synapses. We used a biophysically realistic computational model to examine mechanistically how cholinergic activation of OLMa2 interneurons increases SC to CA1 transmission. Our results suggest that, when properly timed, activation of OLMa2 interneurons cancels the feedforward inhibition onto CA1 pyramidal cells by inhibiting fast-spiking interneurons that synapse on the same dendritic compartment as the SC, i.e., by disinhibiting the pyramidal cell dendritic compartment. Our work further describes the pairing of disinhibition with SC stimulation as a general mechanism for the induction of synaptic plasticity. We found that locally-reduced GABA release (disinhibition) paired with SC stimulation could lead to increased NMDAR activation and intracellular calcium concentration sufficient to upregulate AMPAR permeability and potentiate the excitatory synapse. Our work suggests that inhibitory synapses critically modulate excitatory neurotransmission and induction of plasticity at excitatory synapses. Our work also shows how cholinergic action on OLM interneurons, a mechanism whose disruption is associated with memory impairment, can down-regulate the GABAergic signaling into CA1 pyramidal cells and facilitate potentiation of the SC-CA1 synapse.
Microbiome and behaviour: Exploring underlying mechanisms
Environmental insults alter brain function and behaviour inboth rodents and people. One putative underlying mechanism that has receivedsubstantial attention recently is the gut microbiota, the ecosystem ofsymbiotic microorganisms that populate the intestinal tract, which is known toplay a role in brain health and function via the gut-brain axis. Two keyenvironmental insults known to affect both brain function and behaviour, andthe gut microbiome, are poor diet and psychological stress. While there isstrong evidence for interactions between the microbiome and host physiology inthe context of chronic stress, little is known about the role of the microbiomein the host response to acute stress. Determining the underlying mechanisms bywhich stress may provoke functional changes in the gut and brain is criticalfor developing therapeutics to alleviate adverse consequences of traumaticstress.
Dynamics of the mouse auditory cortex and the perception of sound
Communication between the brain and the gut: Learnings from C. elegans
Sleep and the gut
Sleep is generally associated with the brain but poor sleep impacts the entire body - many diseases are caused or exacerbated by sleep loss. Our work is uncovering ways in which sleep and the body interact. We found a special, two-way relationship between sleep and the gut: the gut is uniquely impacted by sleep loss, and it actively controls sleep quality. These findings could help us understand the origins of sleep as well as develop strategies to offset the negative consequences of inadequate sleep.
Gut Feelings: The Microbiota-Gut-Brain Axis Across the Lifespan
The microbiota-gut-brain axis is emerging as a research area of increasing interest for those investigating the biological and physiological basis of brain development and behaviour during early life, adolescence & ageing. The routes of communication between the gut and brain include the vagus nerve, the immune system, tryptophan metabolism, via the enteric nervous system or by way of microbial metabolites such as short chain fatty acids. Studies in animal models have shown that the development of an appropriate stress response is dependent on the microbiota. Developmentally, a variety of factors can impact the microbiota in early life including mode of birth delivery, antibiotic exposure, mode of nutritional provision, infection, stress as well as host genetics. Recently, the gut microbiota has been implicated in regulating the stress response, and social behaviour. Moreover, fundamental brain processes from adult hippocampal neurogenesis to myelination to microglia activation have been shown to be regulated by the microbiome. Further studies will focus on understanding the mechanisms underlying such brain effects and how they can be exploited by microbiota-targeted interventions including ‘psychobiotics’ and diet
New Strategies and Approaches to Tackle and Understand Neurological Disorder
Broadly, the Mauro Costa-Mattioli laboratory (The MCM Lab) encompasses two complementary lines of research. The first one, more traditional but very important, aims at unraveling the molecular mechanisms underlying memory formation (e.g., using state-of-the-art molecular and cell-specific genetic approaches). Learning and memory disorders can strike the brain during development (e.g., Autism Spectrum Disorders and Down Syndrome), as well as during adulthood (e.g., Alzheimer’s disease). We are interested in understanding the specific circuits and molecular pathways that are primarily targeted in these disorders and how they can be restored. To tackle these questions, we use a multidisciplinary, convergent and cross-species approach that combines mouse and fly genetics, molecular biology, electrophysiology, stem cell biology, optogenetics and behavioral techniques. The second line of research, more recent and relatively unexplored, is focused on understanding how gut microbes control CNS driven-behavior and brain function. Our recent discoveries, that microbes in the gut could modulate brain function and behavior in a very powerful way, have added a whole new dimension to the classic view of how complex behaviors are controlled. The unexpected findings have opened new avenues of study for us and are currently driving my lab to answer a host of new and very interesting questions: - What are the gut microbes (and metabolites) that regulate CNS-driven behaviors? Would it be possible to develop an unbiased screening method to identify specific microbes that regulate different behaviors? - If this is the case, can we identify how members of the gut microbiome (and their metabolites) mechanistically influence brain function? - What is the communication channel between the gut microbiota and the brain? Do different gut microbes use different ways to interact with the brain? - Could disruption of the gut microbial ecology cause neurodevelopmental dysfunction? If so, what is the impact of disruption in young and adult animals? - More importantly, could specific restoration of selected bacterial strains (new generation probiotics) represent a novel therapeutic approach for the targeted treatment of neurodevelopmental disorders? - Finally, can we develop microbiota-directed therapeutic foods to repair brain dysfunction in a variety of neurological disorders?
Early constipation predicts faster dementia onset in Parkinson’s disease
Constipation is a common but not a universal feature in early PD, suggesting that gut involvement is heterogeneous and may be part of a distinct PD subtype with prognostic implications. We analysed data from the Parkinson’s Incidence Cohorts Collaboration, composed of incident community-based cohorts of PD patients assessed longitudinally over 8 years. Constipation was assessed with the MDS-UPDRS constipation item or a comparable categorical scale. Primary PD outcomes of interest were dementia, postural instability and death. PD patients were stratified according to constipation severity at diagnosis: none (n=313, 67.3%), minor (n=97, 20.9%) and major (n=55, 11.8%). Clinical progression to all 3 outcomes was more rapid in those with more severe constipation at baseline (Kaplan Meier survival analysis). Cox regression analysis, adjusting for relevant confounders, confirmed a significant relationship between constipation severity and progression to dementia, but not postural instability or death. Early constipation may predict an accelerated progression of neurodegenerative pathology. Conclusions: We show widespread cortical and subcortical grey matter micro-structure associations with schizophrenia PRS. Across all investigated phenotypes NDI, a measure of the density of myelinated axons and dendrites, showed the most robust associations with schizophrenia PRS. We interpret these results as indicative of reduced density of myelinated axons and dendritic arborization in large-scale cortico-subcortical networks mediating the genetic risk for schizophrenia.
Integration and unification in the science of consciousness
Despite undeniable progress in the science of consciousness, there is no consensus on even fundamental theoretical and empirical questions, such as whether ‘phenomenal consciousness’ is a scientifically respectable concept, whether phenomenal consciousness overflows access consciousness, or whether the neural correlates of perceptual consciousness are in the front or in the back of the cerebral cortex. Notably, disagreement also concerns proposed theories of consciousness. However, since not all theories are mutually incompatible, there have been attempts to make theoretical progress by integrating or unifying them. I shall argue that this is preferable over proposing yet another theory, but that one should not expect it to yield a complete theory of consciousness. Rather, theoretical work in consciousness research should focus on core hypotheses about consciousness that different theories of consciousness have in common. Such a ‘minimal unifying model’ of consciousness can then be used as a basis for formulating more specific hypotheses about consciousness.
Single molecule motion and mixtures: how do human gut bacteria recognize carbohydrates?
Interactions between the microbiome and nervous system during early development
The gut microbiota is emerging as an important modulator of brain function and behavior, as several recent discoveries reveal substantial effects of the microbiome on neurophysiology, neuroimmunity and animal behavior. Despite these findings supporting a “microbiome-gut-brain axis”, the molecular and cellular mechanisms that underlie interactions between the gut microbiota and brain remain poorly understood. To uncover these, the Hsiao laboratory is mining the human microbiota for microbial modulators of host neuroactive molecules, investigating the impact of microbiota-immune system interactions on neurodevelopment and examining the microbiome as an interface between gene-environment interactions in neurological diseases. In particular, our research on effects of the maternal microbiome on offspring development in utero are revealing novel interactions between microbiome-dependent metabolites and fetal thalamocortical axonogenesis. Overall, we aim to dissect biological pathways for communication between the gut microbiota and nervous system, toward understanding fundamental interactions between physiological systems that impact brain and behavior.
A gut choice
Long-term effects of diet-induced obesity on gut-brain communication
Rapid communication between the gut and the brain about recently consumed nutrients is critical for regulating food intake and maintaining energy homeostasis. We have shown that the infusion of nutrients directly into the gastrointestinal tract rapidly inhibits hunger-promoting AgRP neurons in the arcuate nucleus of the hypothalamus and suppresses subsequent feeding. The mechanism of this inhibition appears to be dependent upon macronutrient content, and can be recapitulated by a several hormones secreted in the gut in response to nutrient ingestion. In high-fat diet-induced obese mice, the response of AgRP neurons to nutrient-related stimuli are broadly attenuated. This attenuation is largely irreversible following weight loss and may represent a mechanism underlying difficulty with weight loss and propensity for weight regain in obesity.
What can the gut nervous system tell us about our brain?
Modulation of C. elegans behavior by gut microbes
We are interested in understanding how microbes impact the behavior of host animals. Animal nervous systems likely evolved in environments richly surrounded by microbes, yet the impact of bacteria on nervous system function has been relatively under-studied. A challenge has been to identify systems in which both host and microbe are amenable to genetic manipulation, and which enable high-throughput behavioral screening in response to defined and naturalistic conditions. To accomplish these goals, we use an animal host — the roundworm C. elegans, which feeds on bacteria — in combination with its natural gut microbiome to identify inter-organismal signals driving host-microbe interactions and decision-making. C. elegans has some of the most extensive molecular, neurobiological and genetic tools of any multicellular eukaryote, and, coupled with the ease of gnotobiotic culture in these worms, represents a highly attractive system in which to study microbial influence on host behavior. Using this system, we discovered that commensal bacterial metabolites directly modulate nervous system function of their host. Beneficial gut microbes of the genus Providencia produce the neuromodulator tyramine in the C. elegans intestine. Using a combination of behavioral analysis, neurogenetics, metabolomics and bacterial genetics we established that bacterially produced tyramine is converted to octopamine in C. elegans, which acts directly in sensory neurons to reduce odor aversion and increase sensory preference for Providencia. We think that this type of sensory modulation may increase association of C. elegans with these microbes, increasing availability of this nutrient-rich food source for the worm and its progeny, while facilitating dispersal of the bacteria.
Untitled Seminar
Neuro-immune interactions in pain and host defense
The Chiu laboratory focuses on neuro-immune interactions in pain, itch, and tissue inflammation. Dr. Chiu’s research has uncovered molecular interactions between the nervous system, the immune system and microbes that modulates host defense. He has found that sensory neurons can directly detect bacterial pathogens and their toxins to produce pain. Neurons in turn release neuropeptides that modulate immune cells in host defense. These interactions occur at major tissue barriers in the body including the gut, skin and lungs. In this talk, he will discuss these major neuro-immune interactions and how understanding them could lead to novel approaches to treat pain or inflammation.
Flow, fluctuate and freeze: Epithelial cell sheets as soft active matter
Epithelial cell sheets form a fundamental role in the developing embryo, and also in adult tissues including the gut and the cornea of the eye. Soft and active matter provides a theoretical and computational framework to understand the mechanics and dynamics of these tissues.I will start by introducing the simplest useful class of models, active brownian particles (ABPs), which incorporate uncoordinated active crawling over a substrate and mechanical interactions. Using this model, I will show how the extended ’swirly’ velocity fluctuations seen in sheets on a substrate can be understood using a simple model that couples linear elasticity with disordered activity. We are able to quantitatively match experiments using in-vitro corneal epithelial cells.Adding a different source of activity, cell division and apoptosis, to such a model leads to a novel 'self-melting' dense fluid state. Finally, I will discuss a direct application of this simple particle-based model to the steady-state spiral flow pattern on the mouse cornea.
Sex, guts and babies: the plasticity of the adult intestine and its neurons
Internal organs constantly exchange signals, and can respond with striking anatomical and functional transformations, even in fully developed organisms. We are exploring the mechanisms that drive and sustain such plasticity using the intestine and its neurons as experimental systems. I will present some of our recent work, which has characterised the enteric nervous system of Drosophila, and has explored its physiological plasticity as well as that of the intestine itself. This work has uncovered unexpected sexual dimorphisms, intestinal contributions to reproductive success and metabolic crosstalk between the gut and the brain. Interestingly, this crosstalk appears to be spatially constrained by the three dimensional arrangement of viscera, revealing a previously unrecognised layer of inter-organ signalling regulation. I may also describe our attempts to explore how broadly applicable our findings may be using mammalian systems.
Dynamics of microbiota communities during physical perturbation
The consortium of microbes living in and on our bodies is intimately connected with human biology and deeply influenced by physical forces. Despite incredible gains in describing this community, and emerging knowledge of the mechanisms linking it to human health, understanding the basic physical properties and responses of this ecosystem has been comparatively neglected. Most diseases have significant physical effects on the gut; diarrhea alters osmolality, fever and cancer increase temperature, and bowel diseases affect pH. Furthermore, the gut itself is comprised of localized niches that differ significantly in their physical environment, and are inhabited by different commensal microbes. Understanding the impact of common physical factors is necessary for engineering robust microbiota members and communities; however, our knowledge of how they affect the gut ecosystem is poor. We are investigating how changes in osmolality affect the host and the microbial community and lead to mechanical shifts in the cellular environment. Osmotic perturbation is extremely prevalent in humans, caused by the use of laxatives, lactose intolerance, or celiac disease. In our studies we monitored osmotic shock to the microbiota using a comprehensive and novel approach, which combined in vivo experiments to imaging, physical measurements, computational analysis and highly controlled microfluidic experiments. By bridging several disciplines, we developed a mechanistic understanding of the processes involved in osmotic diarrhea, linking single-cell biophysical changes to large-scale community dynamics. Our results indicate that physical perturbations can profoundly and permanently change the competitive and ecological landscape of the gut, and affect the cell wall of bacteria differentially, depending on their mechanical characteristics.
Food Mind Control: Regulation of Sensory Behaviors by Gut-Brain Signaling
How does the presence or absence of food shape and prioritize behavioral decisions? When is food more than just food? As in other animals, prolonged food deprivation dramatically alters sensory behaviors in C. elegans. For instance, it has been known since the mid-1970s that hungry worms no longer respond to temperature changes in their environment, but the underlying mechanisms have been unclear. I will describe unpublished work showing that insulin signaling from the gut regulates thermosensory behaviors as a function of feeding state by engaging a modulatory sensorimotor circuit that gates the output of the core thermosensory network. C. elegans is associated with, and consumes, diverse bacteria in the wild. I will also discuss a recent story in which we find that in addition to providing nutrition, a bacterial strain in the worm gut alters the hosts’ olfactory behavior and drives food choice decisions by producing a neurotransmitter that targets the hosts’ sensory neurons. These results add to our growing body of knowledge of how signaling from the gut modulates peripheral and central neuron properties and drives sensory behavioral plasticity.
Vision in dynamically changing environments
Many visual systems can process information in dynamically changing environments. In general, visual perception scales with changes in the visual stimulus, or contrast, irrespective of background illumination. This is achieved by adaptation. However, visual perception is challenged when adaptation is not fast enough to deal with sudden changes in overall illumination, for example when gaze follows a moving object from bright sunlight into a shaded area. We have recently shown that the visual system of the fly found a solution by propagating a corrective luminance-sensitive signal to higher processing stages. Using in vivo two-photon imaging and behavioural analyses we showed that distinct OFF-pathway inputs encode contrast and luminance. The luminance-sensitive pathway is particularly required when processing visual motion in contextual dim light, when pure contrast sensitivity underestimates the salience of a stimulus. Recent work in the lab has addressed the question how two visual pathways obtain such fundamentally different sensitivities, given common photoreceptor input. We are furthermore currently working out the network-based strategies by which luminance- and contrast-sensitive signals are combined to guide appropriate visual behaviour. Together, I will discuss the molecular, cellular, and circuit mechanisms that ensure contrast computation, and therefore robust vision, in fast changing visual scenes.
AAV-mediated overexpression of wild-type human alpha-synuclein leads to alterations in gut microbiota in a ‘brain-first’ rat model of prodromal Parkinson’s disease
FENS Forum 2024
Acute stress, microbial metabolites and the microbiota-gut-brain axis: Focus on microbial regulation of barrier function and hippocampal plasticity
FENS Forum 2024
Administration of Enterococcus faecium L-3 reduces disease severity in EAE model in rats by modulating microbiota composition, gut micromorphology, and immune function
FENS Forum 2024
Ameliorative effects of Enterococcus faecium on the gut-brain axis in Parkinson's disease
FENS Forum 2024
APOE genotype effects on the human gut microbiome
FENS Forum 2024
Gut bacterial toxin further enhances blood-brain barrier alterations in a progressive mouse model of Parkinson’s disease
FENS Forum 2024
The brain-gut axis in Alzheimer’s disease: Insights into a new clearance mechanism of amyloid beta peptide and tau protein
FENS Forum 2024
Entorhinal cortex as a hub in the gut microbiome–brain axis
FENS Forum 2024
Exploration of interventions that modulate stroke via gut-brain axis: A meta-analysis
FENS Forum 2024
Extracellular vesicles from mesenchymal stem cells alter gut microbiota and improve neuroinflammation and motor impairment in rats with mild liver damage
FENS Forum 2024
Functional mapping of brain pathways involved in the gut microbial modulation of social behaviour
FENS Forum 2024
The gut-brain axis in an animal model of schizophrenia
FENS Forum 2024
The gut-brain vagal axis governs mesolimbic natural and recreational reward dynamics
FENS Forum 2024
Happy Brain – Happy Bacteria? The effect of electroconvulsive therapy on gut bacteria
FENS Forum 2024
Investigating gut-microbe interactions and epithelial α-synuclein through human enteroid monolayers and imaging flow cytometry of enteroendocrine cells in vitro
FENS Forum 2024
Lactobacillus sakei improves behavioral deficits in Parkinson's disease through the gut-brain axis
FENS Forum 2024
The maternal gut microbiota regulates embryonic cortical development in mice
FENS Forum 2024
Melatonin reduces neuroinflammation and ameliorates gut dysbiosis in the preclinical mouse model of progressive multiple sclerosis
FENS Forum 2024
Method for 3D quantitative analysis of enteric nervous system remodeling in mouse and human gut tissues
FENS Forum 2024
Gut microbiome metabolites and Alzheimer’s disease
FENS Forum 2024
Gut microbiota alterations and hypothalamic inflammation precede obesity in a rat model of binge eating
FENS Forum 2024
Modification of cognitive development in offspring born to pregnant dams with IBD-like gut pathology induced at mid-gestation
FENS Forum 2024
Navigating the challenges of investigating the influence of gestational gut microbiome disruption and perinatal asphyxia on neurodevelopmental reflexes in rat offspring
FENS Forum 2024
A novel sEH inhibitor reduces inflammation and promotes neuroprotective effects by modulating gut microbiota
FENS Forum 2024
An obesity-associated switch in vagal gut-brain communication modulates feeding behavior
FENS Forum 2024
Perinatal methyl donor deficiency increases the prevalence of “depressive-like” behavior in association with alteration of the microbiota-gut-brain dialogue in a transgenerational rat model
FENS Forum 2024
The pesticide glyphosate induces sex-dependent behavioural changes in mice: A role for the gut microbiota?
FENS Forum 2024
Physical activity sensitizes vagal gut-brain communication underlying feeding control
FENS Forum 2024
A preclinical study to explore the glycotoxic impact of methylglyoxal on brain and gut health: Implications for Alzheimer's disease
FENS Forum 2024
Primary sensory neurons require a functional interleukin-6 signal transducer to regulate gut microbiota composition in mice
FENS Forum 2024
Prodromal gut dysbiosis, anhedonia and depression-like behavior in the A53T mouse model of Parkinson’s disease – the impact of social microenvironment
FENS Forum 2024
Role of the gestational maternal gut-microbiota in the neurodevelopment of the hypothalamus and the amygdala
FENS Forum 2024
TAAR1: Bridging ketogenic diet-induced neuroprotection and gut-brain axis in ischemic stroke
FENS Forum 2024
Targeting the gut microbiota to ameliorate the effects of an early-life high-fat/high-sugar diet on eating behaviour in adolescence and adulthood
FENS Forum 2024
Understanding the influence of the gut microbiome on the mesolimbic system and its response to nicotine
FENS Forum 2024