Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130.467
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 42(1): 489-519, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38941607

ABSTRACT

Recent advances have contributed to a mechanistic understanding of neuroimmune interactions in the intestine and revealed an essential role of this cross talk for gut homeostasis and modulation of inflammatory and infectious intestinal diseases. In this review, we describe the innervation of the intestine by intrinsic and extrinsic neurons and then focus on the bidirectional communication between neurons and immune cells. First, we highlight the contribution of neuronal subtypes to the development of colitis and discuss the different immune and epithelial cell types that are regulated by neurons via the release of neuropeptides and neurotransmitters. Next, we review the role of intestinal inflammation in the development of visceral hypersensitivity and summarize how inflammatory mediators induce peripheral and central sensitization of gut-innervating sensory neurons. Finally, we outline the importance of immune cells and gut microbiota for the survival and function of different neuronal populations at homeostasis and during bacterial and helminth infection.


Subject(s)
Neuroimmunomodulation , Humans , Animals , Intestines/immunology , Homeostasis , Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Neurons/metabolism , Neurons/immunology , Neuropeptides/metabolism , Enteric Nervous System/immunology , Enteric Nervous System/metabolism
2.
Annu Rev Immunol ; 40: 143-167, 2022 04 26.
Article in English | MEDLINE | ID: mdl-34990209

ABSTRACT

The gut microbiome influences many host physiologies, spanning gastrointestinal function, metabolism, immune homeostasis, neuroactivity, and behavior. Many microbial effects on the host are orchestrated by bidirectional interactions between the microbiome and immune system. Imbalances in this dialogue can lead to immune dysfunction and immune-mediated conditions in distal organs including the brain. Dysbiosis of the gut microbiome and dysregulated neuroimmune responses are common comorbidities of neurodevelopmental, neuropsychiatric, and neurological disorders, highlighting the importance of the gut microbiome-neuroimmune axis as a regulator of central nervous system homeostasis. In this review, we discuss recent evidence supporting a role for the gut microbiome in regulating the neuroimmune landscape in health and disease.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Animals , Brain , Dysbiosis , Humans , Neuroimmunomodulation
3.
Annu Rev Immunol ; 37: 73-95, 2019 04 26.
Article in English | MEDLINE | ID: mdl-31026414

ABSTRACT

Neurotropic RNA viruses continue to emerge and are increasingly linked to diseases of the central nervous system (CNS) despite viral clearance. Indeed, the overall mortality of viral encephalitis in immunocompetent individuals is low, suggesting efficient mechanisms of virologic control within the CNS. Both immune and neural cells participate in this process, which requires extensive innate immune signaling between resident and infiltrating cells, including microglia and monocytes, that regulate the effector functions of antiviral T and B cells as they gain access to CNS compartments. While these interactions promote viral clearance via mainly neuroprotective mechanisms, they may also promote neuropathology and, in some cases, induce persistent alterations in CNS physiology and function that manifest as neurologic and psychiatric diseases. This review discusses mechanisms of RNA virus clearance and neurotoxicity during viral encephalitis with a focus on the cytokines essential for immune and neural cell inflammatory responses and interactions. Understanding neuroimmune communications in the setting of viral infections is essential for the development of treatments that augment neuroprotective processes while limiting ongoing immunopathological processes that cause ongoing CNS disease.


Subject(s)
Brain/immunology , Immunity, Innate , Microglia/physiology , RNA Virus Infections/immunology , RNA Viruses/physiology , Animals , Blood-Brain Barrier , Brain/virology , Humans , Neurogenic Inflammation , Neuroimmunomodulation
4.
Annu Rev Immunol ; 36: 783-812, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29677475

ABSTRACT

The nervous system regulates immunity and inflammation. The molecular detection of pathogen fragments, cytokines, and other immune molecules by sensory neurons generates immunoregulatory responses through efferent autonomic neuron signaling. The functional organization of this neural control is based on principles of reflex regulation. Reflexes involving the vagus nerve and other nerves have been therapeutically explored in models of inflammatory and autoimmune conditions, and recently in clinical settings. The brain integrates neuro-immune communication, and brain function is altered in diseases characterized by peripheral immune dysregulation and inflammation. Here we review the anatomical and molecular basis of the neural interface with immunity, focusing on peripheral neural control of immune functions and the role of the brain in the model of the immunological homunculus. Clinical advances stemming from this knowledge within the framework of bioelectronic medicine are also briefly outlined.


Subject(s)
Neuroimmunomodulation , Animals , Biomarkers , Disease Susceptibility , Humans , Immunity , Nervous System/anatomy & histology , Nervous System/immunology , Nervous System/metabolism , Nervous System Physiological Phenomena , Neuroimmunomodulation/genetics , Neuroimmunomodulation/immunology , Signal Transduction , Translational Research, Biomedical
5.
Annu Rev Biochem ; 93(1): 339-366, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38346274

ABSTRACT

The nicotinic acetylcholine receptor has served, since its biochemical identification in the 1970s, as a model of an allosteric ligand-gated ion channel mediating signal transition at the synapse. In recent years, the application of X-ray crystallography and high-resolution cryo-electron microscopy, together with molecular dynamic simulations of nicotinic receptors and homologs, have opened a new era in the understanding of channel gating by the neurotransmitter. They reveal, at atomic resolution, the diversity and flexibility of the multiple ligand-binding sites, including recently discovered allosteric modulatory sites distinct from the neurotransmitter orthosteric site, and the conformational dynamics of the activation process as a molecular switch linking these multiple sites. The model emerging from these studies paves the way for a new pharmacology based, first, upon the occurrence of an original mode of indirect allosteric modulation, distinct from a steric competition for a single and rigid binding site, and second, the design of drugs that specifically interact with privileged conformations of the receptor such as agonists, antagonists, and desensitizers. Research on nicotinic receptors is still at the forefront of understanding the mode of action of drugs on the nervous system.


Subject(s)
Allosteric Site , Cryoelectron Microscopy , Molecular Dynamics Simulation , Receptors, Nicotinic , Signal Transduction , Receptors, Nicotinic/metabolism , Receptors, Nicotinic/chemistry , Receptors, Nicotinic/genetics , Allosteric Regulation , Humans , Animals , Crystallography, X-Ray , Binding Sites , Protein Conformation , Ligands , Models, Molecular , Protein Multimerization , Nicotinic Agonists/chemistry , Nicotinic Agonists/pharmacology , Nicotinic Agonists/metabolism
6.
Cell ; 187(17): 4733-4750.e26, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-38971152

ABSTRACT

We identify a population of Protogenin-positive (PRTG+ve) MYChigh NESTINlow stem cells in the four-week-old human embryonic hindbrain that subsequently localizes to the ventricular zone of the rhombic lip (RLVZ). Oncogenic transformation of early Prtg+ve rhombic lip stem cells initiates group 3 medulloblastoma (Gr3-MB)-like tumors. PRTG+ve stem cells grow adjacent to a human-specific interposed vascular plexus in the RLVZ, a phenotype that is recapitulated in Gr3-MB but not in other types of medulloblastoma. Co-culture of Gr3-MB with endothelial cells promotes tumor stem cell growth, with the endothelial cells adopting an immature phenotype. Targeting the PRTGhigh compartment of Gr3-MB in vivo using either the diphtheria toxin system or chimeric antigen receptor T cells constitutes effective therapy. Human Gr3-MBs likely arise from early embryonic RLVZ PRTG+ve stem cells inhabiting a specific perivascular niche. Targeting the PRTGhigh compartment and/or the perivascular niche represents an approach to treat children with Gr3-MB.


Subject(s)
Medulloblastoma , Neoplastic Stem Cells , Humans , Medulloblastoma/pathology , Medulloblastoma/metabolism , Animals , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Mice , Rhombencephalon/metabolism , Rhombencephalon/embryology , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Endothelial Cells/metabolism , Stem Cell Niche , Stem Cells/metabolism , Coculture Techniques , Embryonic Structures , Metencephalon/embryology
7.
Cell ; 187(3): 712-732.e38, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38194967

ABSTRACT

Human brain development involves an orchestrated, massive neural progenitor expansion while a multi-cellular tissue architecture is established. Continuously expanding organoids can be grown directly from multiple somatic tissues, yet to date, brain organoids can solely be established from pluripotent stem cells. Here, we show that healthy human fetal brain in vitro self-organizes into organoids (FeBOs), phenocopying aspects of in vivo cellular heterogeneity and complex organization. FeBOs can be expanded over long time periods. FeBO growth requires maintenance of tissue integrity, which ensures production of a tissue-like extracellular matrix (ECM) niche, ultimately endowing FeBO expansion. FeBO lines derived from different areas of the central nervous system (CNS), including dorsal and ventral forebrain, preserve their regional identity and allow to probe aspects of positional identity. Using CRISPR-Cas9, we showcase the generation of syngeneic mutant FeBO lines for the study of brain cancer. Taken together, FeBOs constitute a complementary CNS organoid platform.


Subject(s)
Brain , Organoids , Humans , Brain/cytology , Brain/growth & development , Brain/metabolism , Central Nervous System/metabolism , Extracellular Matrix/metabolism , Pluripotent Stem Cells/metabolism , Prosencephalon/cytology , Tissue Culture Techniques , Stem Cells/metabolism , Morphogenesis
8.
Cell ; 187(8): 1955-1970.e23, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38503282

ABSTRACT

Characterizing somatic mutations in the brain is important for disentangling the complex mechanisms of aging, yet little is known about mutational patterns in different brain cell types. Here, we performed whole-genome sequencing (WGS) of 86 single oligodendrocytes, 20 mixed glia, and 56 single neurons from neurotypical individuals spanning 0.4-104 years of age and identified >92,000 somatic single-nucleotide variants (sSNVs) and small insertions/deletions (indels). Although both cell types accumulate somatic mutations linearly with age, oligodendrocytes accumulated sSNVs 81% faster than neurons and indels 28% slower than neurons. Correlation of mutations with single-nucleus RNA profiles and chromatin accessibility from the same brains revealed that oligodendrocyte mutations are enriched in inactive genomic regions and are distributed across the genome similarly to mutations in brain cancers. In contrast, neuronal mutations are enriched in open, transcriptionally active chromatin. These stark differences suggest an assortment of active mutagenic processes in oligodendrocytes and neurons.


Subject(s)
Aging , Brain , Neurons , Oligodendroglia , Humans , Aging/genetics , Aging/pathology , Chromatin/genetics , Chromatin/metabolism , Mutation , Neurons/metabolism , Neurons/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Single-Cell Gene Expression Analysis , Whole Genome Sequencing , Brain/metabolism , Brain/pathology , Polymorphism, Single Nucleotide , INDEL Mutation , Biological Specimen Banks , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Precursor Cells/pathology
9.
Cell ; 187(9): 2143-2157.e15, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38670072

ABSTRACT

A central question for regenerative neuroscience is whether synthetic neural circuits, such as those built from two species, can function in an intact brain. Here, we apply blastocyst complementation to selectively build and test interspecies neural circuits. Despite approximately 10-20 million years of evolution, and prominent species differences in brain size, rat pluripotent stem cells injected into mouse blastocysts develop and persist throughout the mouse brain. Unexpectedly, the mouse niche reprograms the birth dates of rat neurons in the cortex and hippocampus, supporting rat-mouse synaptic activity. When mouse olfactory neurons are genetically silenced or killed, rat neurons restore information flow to odor processing circuits. Moreover, they rescue the primal behavior of food seeking, although less well than mouse neurons. By revealing that a mouse can sense the world using neurons from another species, we establish neural blastocyst complementation as a powerful tool to identify conserved mechanisms of brain development, plasticity, and repair.


Subject(s)
Neurons , Animals , Mice , Rats , Neurons/metabolism , Neurons/cytology , Neurons/physiology , Blastocyst/metabolism , Blastocyst/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Brain/cytology , Brain/physiology , Female , Hippocampus/cytology , Hippocampus/physiology , Species Specificity , Mice, Inbred C57BL , Male
10.
Cell ; 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38986619

ABSTRACT

Posterior fossa group A (PFA) ependymoma is a lethal brain cancer diagnosed in infants and young children. The lack of driver events in the PFA linear genome led us to search its 3D genome for characteristic features. Here, we reconstructed 3D genomes from diverse childhood tumor types and uncovered a global topology in PFA that is highly reminiscent of stem and progenitor cells in a variety of human tissues. A remarkable feature exclusively present in PFA are type B ultra long-range interactions in PFAs (TULIPs), regions separated by great distances along the linear genome that interact with each other in the 3D nuclear space with surprising strength. TULIPs occur in all PFA samples and recur at predictable genomic coordinates, and their formation is induced by expression of EZHIP. The universality of TULIPs across PFA samples suggests a conservation of molecular principles that could be exploited therapeutically.

11.
Cell ; 187(10): 2521-2535.e21, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38697107

ABSTRACT

Cancer immunotherapy remains limited by poor antigenicity and a regulatory tumor microenvironment (TME). Here, we create "onion-like" multi-lamellar RNA lipid particle aggregates (LPAs) to substantially enhance the payload packaging and immunogenicity of tumor mRNA antigens. Unlike current mRNA vaccine designs that rely on payload packaging into nanoparticle cores for Toll-like receptor engagement in immune cells, systemically administered RNA-LPAs activate RIG-I in stromal cells, eliciting massive cytokine/chemokine response and dendritic cell/lymphocyte trafficking that provokes cancer immunogenicity and mediates rejection of both early- and late-stage murine tumor models. In client-owned canines with terminal gliomas, RNA-LPAs improved survivorship and reprogrammed the TME, which became "hot" within days of a single infusion. In a first-in-human trial, RNA-LPAs elicited rapid cytokine/chemokine release, immune activation/trafficking, tissue-confirmed pseudoprogression, and glioma-specific immune responses in glioblastoma patients. These data support RNA-LPAs as a new technology that simultaneously reprograms the TME while eliciting rapid and enduring cancer immunotherapy.


Subject(s)
Immunotherapy , Lipids , RNA , Tumor Microenvironment , Animals , Dogs , Female , Humans , Mice , Antigens, Neoplasm/immunology , Brain Neoplasms/therapy , Brain Neoplasms/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Glioblastoma/therapy , Glioblastoma/immunology , Glioma/therapy , Glioma/immunology , Immunotherapy/methods , Mice, Inbred C57BL , Neoplasms/therapy , Neoplasms/immunology , RNA/chemistry , RNA/therapeutic use , RNA, Messenger/metabolism , RNA, Messenger/genetics , Lipids/chemistry
12.
Cell ; 187(13): 3236-3248.e21, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38772369

ABSTRACT

Leveraging AAVs' versatile tropism and labeling capacity, we expanded the scale of in vivo CRISPR screening with single-cell transcriptomic phenotyping across embryonic to adult brains and peripheral nervous systems. Through extensive tests of 86 vectors across AAV serotypes combined with a transposon system, we substantially amplified labeling efficacy and accelerated in vivo gene delivery from weeks to days. Our proof-of-principle in utero screen identified the pleiotropic effects of Foxg1, highlighting its tight regulation of distinct networks essential for cell fate specification of Layer 6 corticothalamic neurons. Notably, our platform can label >6% of cerebral cells, surpassing the current state-of-the-art efficacy at <0.1% by lentivirus, to achieve analysis of over 30,000 cells in one experiment and enable massively parallel in vivo Perturb-seq. Compatible with various phenotypic measurements (single-cell or spatial multi-omics), it presents a flexible approach to interrogate gene function across cell types in vivo, translating gene variants to their causal function.


Subject(s)
Gene Regulatory Networks , Single-Cell Analysis , Animals , Female , Humans , Mice , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , CRISPR-Cas Systems/genetics , Dependovirus/genetics , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Genetic Vectors/metabolism , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/cytology , Single-Cell Analysis/methods , Transcriptome/genetics , Cell Line , Transcription, Genetic
13.
Cell ; 187(3): 676-691.e16, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38306983

ABSTRACT

Behavior relies on activity in structured neural circuits that are distributed across the brain, but most experiments probe neurons in a single area at a time. Using multiple Neuropixels probes, we recorded from multi-regional loops connected to the anterior lateral motor cortex (ALM), a circuit node mediating memory-guided directional licking. Neurons encoding sensory stimuli, choices, and actions were distributed across the brain. However, choice coding was concentrated in the ALM and subcortical areas receiving input from the ALM in an ALM-dependent manner. Diverse orofacial movements were encoded in the hindbrain; midbrain; and, to a lesser extent, forebrain. Choice signals were first detected in the ALM and the midbrain, followed by the thalamus and other brain areas. At movement initiation, choice-selective activity collapsed across the brain, followed by new activity patterns driving specific actions. Our experiments provide the foundation for neural circuit models of decision-making and movement initiation.


Subject(s)
Movement , Neurons , Brain/physiology , Movement/physiology , Neurons/physiology , Thalamus/physiology , Memory
14.
Cell ; 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39178853

ABSTRACT

Animals adapt to environmental conditions by modifying the function of their internal organs, including the brain. To be adaptive, alterations in behavior must be coordinated with the functional state of organs throughout the body. Here, we find that thyroid hormone-a regulator of metabolism in many peripheral organs-directly activates cell-type-specific transcriptional programs in the frontal cortex of adult male mice. These programs are enriched for axon-guidance genes in glutamatergic projection neurons, synaptic regulatory genes in both astrocytes and neurons, and pro-myelination factors in oligodendrocytes, suggesting widespread plasticity of cortical circuits. Indeed, whole-cell electrophysiology revealed that thyroid hormone alters excitatory and inhibitory synaptic transmission, an effect that requires thyroid hormone-induced gene regulatory programs in presynaptic neurons. Furthermore, thyroid hormone action in the frontal cortex regulates innate exploratory behaviors and causally promotes exploratory decision-making. Thus, thyroid hormone acts directly on the cerebral cortex in males to coordinate exploratory behaviors with whole-body metabolic state.

15.
Cell ; 187(2): 446-463.e16, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38242087

ABSTRACT

Treatment failure for the lethal brain tumor glioblastoma (GBM) is attributed to intratumoral heterogeneity and tumor evolution. We utilized 3D neuronavigation during surgical resection to acquire samples representing the whole tumor mapped by 3D spatial coordinates. Integrative tissue and single-cell analysis revealed sources of genomic, epigenomic, and microenvironmental intratumoral heterogeneity and their spatial patterning. By distinguishing tumor-wide molecular features from those with regional specificity, we inferred GBM evolutionary trajectories from neurodevelopmental lineage origins and initiating events such as chromothripsis to emergence of genetic subclones and spatially restricted activation of differential tumor and microenvironmental programs in the core, periphery, and contrast-enhancing regions. Our work depicts GBM evolution and heterogeneity from a 3D whole-tumor perspective, highlights potential therapeutic targets that might circumvent heterogeneity-related failures, and establishes an interactive platform enabling 360° visualization and analysis of 3D spatial patterns for user-selected genes, programs, and other features across whole GBM tumors.


Subject(s)
Brain Neoplasms , Glioblastoma , Models, Biological , Humans , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Epigenomics , Genomics , Glioblastoma/genetics , Glioblastoma/pathology , Single-Cell Analysis , Tumor Microenvironment , Genetic Heterogeneity
16.
Cell ; 187(17): 4637-4655.e26, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39043180

ABSTRACT

The medical burden of stroke extends beyond the brain injury itself and is largely determined by chronic comorbidities that develop secondarily. We hypothesized that these comorbidities might share a common immunological cause, yet chronic effects post-stroke on systemic immunity are underexplored. Here, we identify myeloid innate immune memory as a cause of remote organ dysfunction after stroke. Single-cell sequencing revealed persistent pro-inflammatory changes in monocytes/macrophages in multiple organs up to 3 months after brain injury, notably in the heart, leading to cardiac fibrosis and dysfunction in both mice and stroke patients. IL-1ß was identified as a key driver of epigenetic changes in innate immune memory. These changes could be transplanted to naive mice, inducing cardiac dysfunction. By neutralizing post-stroke IL-1ß or blocking pro-inflammatory monocyte trafficking with a CCR2/5 inhibitor, we prevented post-stroke cardiac dysfunction. Such immune-targeted therapies could potentially prevent various IL-1ß-mediated comorbidities, offering a framework for secondary prevention immunotherapy.


Subject(s)
Brain Injuries , Immunity, Innate , Immunologic Memory , Inflammation , Interleukin-1beta , Mice, Inbred C57BL , Monocytes , Animals , Mice , Interleukin-1beta/metabolism , Brain Injuries/immunology , Humans , Male , Monocytes/metabolism , Monocytes/immunology , Inflammation/immunology , Macrophages/immunology , Macrophages/metabolism , Stroke/complications , Stroke/immunology , Heart Diseases/immunology , Female , Receptors, CCR2/metabolism , Fibrosis , Epigenesis, Genetic , Trained Immunity
17.
Cell ; 186(26): 5892-5909.e22, 2023 12 21.
Article in English | MEDLINE | ID: mdl-38091994

ABSTRACT

Different functional regions of brain are fundamental for basic neurophysiological activities. However, the regional specification remains largely unexplored during human brain development. Here, by combining spatial transcriptomics (scStereo-seq) and scRNA-seq, we built a spatiotemporal developmental atlas of multiple human brain regions from 6-23 gestational weeks (GWs). We discovered that, around GW8, radial glia (RG) cells have displayed regional heterogeneity and specific spatial distribution. Interestingly, we found that the regional heterogeneity of RG subtypes contributed to the subsequent neuronal specification. Specifically, two diencephalon-specific subtypes gave rise to glutamatergic and GABAergic neurons, whereas subtypes in ventral midbrain were associated with the dopaminergic neurons. Similar GABAergic neuronal subtypes were shared between neocortex and diencephalon. Additionally, we revealed that cell-cell interactions between oligodendrocyte precursor cells and GABAergic neurons influenced and promoted neuronal development coupled with regional specification. Altogether, this study provides comprehensive insights into the regional specification in the developing human brain.


Subject(s)
Brain , Transcriptome , Humans , Dopaminergic Neurons , GABAergic Neurons , Mesencephalon , Neocortex , Brain/growth & development , Brain/metabolism
18.
Cell ; 186(12): 2574-2592.e20, 2023 06 08.
Article in English | MEDLINE | ID: mdl-37192620

ABSTRACT

Serotonin influences many aspects of animal behavior. But how serotonin acts on its diverse receptors across the brain to modulate global activity and behavior is unknown. Here, we examine how serotonin release in C. elegans alters brain-wide activity to induce foraging behaviors, like slow locomotion and increased feeding. Comprehensive genetic analyses identify three core serotonin receptors (MOD-1, SER-4, and LGC-50) that induce slow locomotion upon serotonin release and others (SER-1, SER-5, and SER-7) that interact with them to modulate this behavior. SER-4 induces behavioral responses to sudden increases in serotonin release, whereas MOD-1 induces responses to persistent release. Whole-brain imaging reveals widespread serotonin-associated brain dynamics, spanning many behavioral networks. We map all sites of serotonin receptor expression in the connectome, which, together with synaptic connectivity, helps predict which neurons show serotonin-associated activity. These results reveal how serotonin acts at defined sites across a connectome to modulate brain-wide activity and behavior.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans/metabolism , Serotonin/metabolism , Caenorhabditis elegans Proteins/metabolism , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism , Behavior, Animal/physiology , Brain/metabolism
19.
Cell ; 186(2): 398-412.e17, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36669474

ABSTRACT

Public health studies indicate that artificial light is a high-risk factor for metabolic disorders. However, the neural mechanism underlying metabolic modulation by light remains elusive. Here, we found that light can acutely decrease glucose tolerance (GT) in mice by activation of intrinsically photosensitive retinal ganglion cells (ipRGCs) innervating the hypothalamic supraoptic nucleus (SON). Vasopressin neurons in the SON project to the paraventricular nucleus, then to the GABAergic neurons in the solitary tract nucleus, and eventually to brown adipose tissue (BAT). Light activation of this neural circuit directly blocks adaptive thermogenesis in BAT, thereby decreasing GT. In humans, light also modulates GT at the temperature where BAT is active. Thus, our work unveils a retina-SON-BAT axis that mediates the effect of light on glucose metabolism, which may explain the connection between artificial light and metabolic dysregulation, suggesting a potential prevention and treatment strategy for managing glucose metabolic disorders.


Subject(s)
Adipose Tissue, Brown , Hypothalamus , Mice , Animals , Humans , Adipose Tissue, Brown/metabolism , Hypothalamus/metabolism , Thermogenesis/physiology , Retina , Retinal Ganglion Cells , Glucose/metabolism
20.
Cell ; 186(19): 4134-4151.e31, 2023 09 14.
Article in English | MEDLINE | ID: mdl-37607537

ABSTRACT

Changes in an animal's behavior and internal state are accompanied by widespread changes in activity across its brain. However, how neurons across the brain encode behavior and how this is impacted by state is poorly understood. We recorded brain-wide activity and the diverse motor programs of freely moving C. elegans and built probabilistic models that explain how each neuron encodes quantitative behavioral features. By determining the identities of the recorded neurons, we created an atlas of how the defined neuron classes in the C. elegans connectome encode behavior. Many neuron classes have conjunctive representations of multiple behaviors. Moreover, although many neurons encode current motor actions, others integrate recent actions. Changes in behavioral state are accompanied by widespread changes in how neurons encode behavior, and we identify these flexible nodes in the connectome. Our results provide a global map of how the cell types across an animal's brain encode its behavior.


Subject(s)
Caenorhabditis elegans , Connectome , Animals , Brain/cytology , Brain/metabolism , Models, Statistical , Neurons/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL