51
|
Mountoufaris G, Nair A, Yang B, Kim DW, Anderson DJ. Neuropeptide Signaling is Required to Implement a Line Attractor Encoding a Persistent Internal Behavioral State. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.01.565073. [PMID: 37961374 PMCID: PMC10635056 DOI: 10.1101/2023.11.01.565073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Internal states drive survival behaviors, but their neural implementation is not well understood. Recently we identified a line attractor in the ventromedial hypothalamus (VMH) that represents an internal state of aggressiveness. Line attractors can be implemented by recurrent connectivity and/or neuromodulatory signaling, but evidence for the latter is scant. Here we show that neuropeptidergic signaling is necessary for line attractor dynamics in this system, using a novel approach that integrates cell type-specific, anatomically restricted CRISPR/Cas9-based gene editing with microendoscopic calcium imaging. Co-disruption of receptors for oxytocin and vasopressin in adult VMH Esr1 + neurons that control aggression suppressed attack, reduced persistent neural activity and eliminated line attractor dynamics, while only modestly impacting neural activity and sex- or behavior-tuning. These data identify a requisite role for neuropeptidergic signaling in implementing a behaviorally relevant line attractor. Our approach should facilitate mechanistic studies in neuroscience that bridge different levels of biological function and abstraction.
Collapse
|
52
|
Abstract
The nervous system coordinates various motivated behaviors such as feeding, drinking, and escape to promote survival and evolutionary fitness. Although the precise behavioral repertoires required for distinct motivated behaviors are diverse, common features such as approach or avoidance suggest that common brain substrates are required for a wide range of motivated behaviors. In this Review, I describe a framework by which neural circuits specified for some innate drives regulate the activity of ventral tegmental area (VTA) dopamine neurons to reinforce ongoing or planned actions to fulfill motivational demands. This framework may explain why signaling from VTA dopamine neurons is ubiquitously involved in many types of diverse volitional motivated actions, as well as how sensory and interoceptive cues can initiate specific goal-directed actions.
Collapse
Affiliation(s)
- Garret D Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
53
|
Sayar-Atasoy N, Laule C, Aklan I, Kim H, Yavuz Y, Ates T, Coban I, Koksalar-Alkan F, Rysted J, Davis D, Singh U, Alp MI, Yilmaz B, Cui H, Atasoy D. Adrenergic modulation of melanocortin pathway by hunger signals. Nat Commun 2023; 14:6602. [PMID: 37857606 PMCID: PMC10587058 DOI: 10.1038/s41467-023-42362-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 10/09/2023] [Indexed: 10/21/2023] Open
Abstract
Norepinephrine (NE) is a well-known appetite regulator, and the nor/adrenergic system is targeted by several anti-obesity drugs. To better understand the circuitry underlying adrenergic appetite control, here we investigated the paraventricular hypothalamic nucleus (PVN), a key brain region that integrates energy signals and receives dense nor/adrenergic input, using a mouse model. We found that PVN NE level increases with signals of energy deficit and decreases with food access. This pattern is recapitulated by the innervating catecholaminergic axon terminals originating from NTSTH-neurons. Optogenetic activation of rostral-NTSTH → PVN projection elicited strong motivation to eat comparable to overnight fasting whereas its inhibition attenuated both fasting-induced & hypoglycemic feeding. We found that NTSTH-axons functionally targeted PVNMC4R-neurons by predominantly inhibiting them, in part, through α1-AR mediated potentiation of GABA release from ARCAgRP presynaptic terminals. Furthermore, glucoprivation suppressed PVNMC4R activity, which was required for hypoglycemic feeding response. These results define an ascending nor/adrenergic circuit, NTSTH → PVNMC4R, that conveys peripheral hunger signals to melanocortin pathway.
Collapse
Affiliation(s)
- Nilufer Sayar-Atasoy
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Connor Laule
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Iltan Aklan
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Hyojin Kim
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Yavuz Yavuz
- Department of Physiology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Tayfun Ates
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Ilknur Coban
- Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | | | - Jacob Rysted
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Debbie Davis
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Uday Singh
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Muhammed Ikbal Alp
- Department of Physiology, School of Medicine, Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Bayram Yilmaz
- Department of Physiology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Huxing Cui
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Deniz Atasoy
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
54
|
Fernandez G, De Francesco PN, Cornejo MP, Cabral A, Aguggia JP, Duque VJ, Sayar N, Cantel S, Burgos JI, Fehrentz JA, Rorato R, Atasoy D, Mecawi AS, Perello M. Ghrelin Action in the PVH of Male Mice: Accessibility, Neuronal Targets, and CRH Neurons Activation. Endocrinology 2023; 164:bqad154. [PMID: 37823477 PMCID: PMC11491828 DOI: 10.1210/endocr/bqad154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/08/2023] [Accepted: 10/11/2023] [Indexed: 10/13/2023]
Abstract
The hormone ghrelin displays several well-characterized functions, including some with pharmaceutical interest. The receptor for ghrelin, the growth hormone secretagogue receptor (GHSR), is expressed in the hypothalamic paraventricular nucleus (PVH), a critical hub for the integration of metabolic, neuroendocrine, autonomic, and behavioral functions. Here, we performed a neuroanatomical and functional characterization of the neuronal types mediating ghrelin actions in the PVH of male mice. We found that fluorescent ghrelin mainly labels PVH neurons immunoreactive for nitric oxide synthase 1 (NOS1), which catalyze the production of nitric oxide [NO]). Centrally injected ghrelin increases c-Fos in NOS1 PVH neurons and NOS1 phosphorylation in the PVH. We also found that a high dose of systemically injected ghrelin increases the ghrelin level in the cerebrospinal fluid and in the periventricular PVH, and induces c-Fos in NOS1 PVH neurons. Such a high dose of systemically injected ghrelin activates a subset of NOS1 PVH neurons, which do not express oxytocin, via an arcuate nucleus-independent mechanism. Finally, we found that pharmacological inhibition of NO production fully abrogates ghrelin-induced increase of calcium concentration in corticotropin-releasing hormone neurons of the PVH whereas it partially impairs ghrelin-induced increase of plasma glucocorticoid levels. Thus, plasma ghrelin can directly target a subset of NO-producing neurons of the PVH that is involved in ghrelin-induced activation of the hypothalamic-pituitary-adrenal neuroendocrine axis.
Collapse
Affiliation(s)
- Gimena Fernandez
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Pablo N De Francesco
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - María P Cornejo
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Agustina Cabral
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Julieta P Aguggia
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Victor J Duque
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, CEP: 04023-062, Brazil
| | - Nilufer Sayar
- Department of Neuroscience and Pharmacology, Carver College of Medicine, Iowa Neuroscience Institute and Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa, Iowa City, IA 52242, USA
| | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, University of Montpellier, CNRS, ENSCM, Montpellier cedex 5 34293, France
| | - Juan I Burgos
- Centro de Investigaciones Cardiovasculares “Dr. Horacio Eugenio Cingolani” (CONICET and National University of La Plata), La Plata 1900, Buenos Aires, Argentina
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, University of Montpellier, CNRS, ENSCM, Montpellier cedex 5 34293, France
| | - Rodrigo Rorato
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, CEP: 04023-062, Brazil
| | - Deniz Atasoy
- Department of Neuroscience and Pharmacology, Carver College of Medicine, Iowa Neuroscience Institute and Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa, Iowa City, IA 52242, USA
| | - André S Mecawi
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, CEP: 04023-062, Brazil
| | - Mario Perello
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala 751 05, Sweden
| |
Collapse
|
55
|
Ma L, Liu Q, Liu X, Chang H, Jin S, Ma W, Xu F, Liu H. Paraventricular Hypothalamic Nucleus Upregulates Intraocular Pressure Via Glutamatergic Neurons. Invest Ophthalmol Vis Sci 2023; 64:43. [PMID: 37773501 PMCID: PMC10547014 DOI: 10.1167/iovs.64.12.43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 08/11/2023] [Indexed: 10/01/2023] Open
Abstract
Purpose The neuroregulatory center of intraocular pressure (IOP) is located in the hypothalamus. An efferent neural pathway exists between the hypothalamic nuclei and the autonomic nerve endings in the anterior chamber of the eye. This study was designed to investigate whether the paraventricular hypothalamic nucleus (PVH) regulates IOP as the other nuclei do. Methods Optogenetic manipulation of PVH neurons was used in this study. Light stimulation was applied via an optical fiber embedded over the PVH to activate projection neurons after AAV2/9-CaMKIIα-hChR2-mCherry was injected into the right PVH of C57BL/6J mice. The same methods were used to inhibit projection neurons after AAV2/9-CaMKIIα-eNpHR3.0-mCherry was injected into the bilateral PVH of C57BL/6J mice. AAV2/9-EF1α-DIO-hChR2-mCherry was injected into the right PVH of Vglut2-Cre mice to elucidate the effect of glutamatergic neuron-specific activation. IOP was measured before and after light manipulation. Associated nuclei activation was clarified by c-Fos immunohistochemical staining. Only mice with accurate viral expression and fiber embedding were included in the statistical analysis. Results Activation of projection neurons in the right PVH induced significant bilateral IOP elevation (n = 11, P < 0.001); the ipsilateral IOP increased more noticeably (n = 11, P < 0.05); Bilateral inhibition of PVH projection neurons did not significantly influence IOP (n = 5, P > 0.05). Specific activation of glutamatergic neurons among PVH projection neurons also induced IOP elevation in both eyes (n = 5, P < 0.001). The dorsomedial hypothalamic nucleus, ventromedial hypothalamic nucleus, locus coeruleus and basolateral amygdaloid nucleus responded to light stimulation of PVH in AAV-ChR2 mice. Conclusions The PVH may play a role in IOP upregulation via glutamatergic neurons.
Collapse
Affiliation(s)
- Lin Ma
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Liu
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xin Liu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Chang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sen Jin
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Wenyu Ma
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Fuqiang Xu
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Haixia Liu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
56
|
Douglass AM, Resch JM, Madara JC, Kucukdereli H, Yizhar O, Grama A, Yamagata M, Yang Z, Lowell BB. Neural basis for fasting activation of the hypothalamic-pituitary-adrenal axis. Nature 2023; 620:154-162. [PMID: 37495689 PMCID: PMC11168300 DOI: 10.1038/s41586-023-06358-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 06/21/2023] [Indexed: 07/28/2023]
Abstract
Fasting initiates a multitude of adaptations to allow survival. Activation of the hypothalamic-pituitary-adrenal (HPA) axis and subsequent release of glucocorticoid hormones is a key response that mobilizes fuel stores to meet energy demands1-5. Despite the importance of the HPA axis response, the neural mechanisms that drive its activation during energy deficit are unknown. Here, we show that fasting-activated hypothalamic agouti-related peptide (AgRP)-expressing neurons trigger and are essential for fasting-induced HPA axis activation. AgRP neurons do so through projections to the paraventricular hypothalamus (PVH), where, in a mechanism not previously described for AgRP neurons, they presynaptically inhibit the terminals of tonically active GABAergic afferents from the bed nucleus of the stria terminalis (BNST) that otherwise restrain activity of corticotrophin-releasing hormone (CRH)-expressing neurons. This disinhibition of PVHCrh neurons requires γ-aminobutyric acid (GABA)/GABA-B receptor signalling and potently activates the HPA axis. Notably, stimulation of the HPA axis by AgRP neurons is independent of their induction of hunger, showing that these canonical 'hunger neurons' drive many distinctly different adaptations to the fasted state. Together, our findings identify the neural basis for fasting-induced HPA axis activation and uncover a unique means by which AgRP neurons activate downstream neurons: through presynaptic inhibition of GABAergic afferents. Given the potency of this disinhibition of tonically active BNST afferents, other activators of the HPA axis, such as psychological stress, may also work by reducing BNST inhibitory tone onto PVHCrh neurons.
Collapse
Affiliation(s)
- Amelia M Douglass
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jon M Resch
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Joseph C Madara
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Hakan Kucukdereli
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ofer Yizhar
- Departments of Brain Sciences and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Abhinav Grama
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Masahito Yamagata
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Zongfang Yang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Program in Neuroscience, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
57
|
Shin MG, Bae Y, Afzal R, Kondoh K, Lee EJ. Olfactory modulation of stress-response neural circuits. Exp Mol Med 2023; 55:1659-1671. [PMID: 37524867 PMCID: PMC10474124 DOI: 10.1038/s12276-023-01048-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/20/2023] [Accepted: 04/30/2023] [Indexed: 08/02/2023] Open
Abstract
Stress responses, which are crucial for survival, are evolutionally conserved throughout the animal kingdom. The most common endocrine axis among stress responses is that triggered by corticotropin-releasing hormone neurons (CRHNs) in the hypothalamus. Signals of various stressors are detected by different sensory systems and relayed through individual neural circuits that converge on hypothalamic CRHNs to initiate common stress hormone responses. To investigate the neurocircuitry mechanisms underlying stress hormone responses induced by a variety of stressors, researchers have recently developed new approaches employing retrograde transsynaptic viral tracers, providing a wealth of information about various types of neural circuits that control the activity of CRHNs in response to stress stimuli. Here, we review earlier and more recent findings on the stress neurocircuits that converge on CRHNs, focusing particularly on olfactory systems that excite or suppress the activities of CRHNs and lead to the initiation of stress responses. Because smells are arguably the most important signals that enable animals to properly cope with environmental changes and survive, unveiling the regulatory mechanisms by which smells control stress responses would provide broad insight into how stress-related environmental cues are perceived in the animal brain.
Collapse
Affiliation(s)
- Min-Gi Shin
- Department of Brain Science, Ajou University School of Medicine, Suwon, 16499, Korea
- AI-Superconvergence KIURI Translational Research Center, Ajou University School of Medicine, Suwon, 16499, Korea
| | - Yiseul Bae
- Department of Brain Science, Ajou University School of Medicine, Suwon, 16499, Korea
| | - Ramsha Afzal
- Department of Brain Science, Ajou University School of Medicine, Suwon, 16499, Korea
| | - Kunio Kondoh
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi, 444-8585, Japan.
- Japan Science and Technology Agency, PRESTO, Okazaki, Aichi, 444-8585, Japan.
| | - Eun Jeong Lee
- Department of Brain Science, Ajou University School of Medicine, Suwon, 16499, Korea.
| |
Collapse
|
58
|
Zhang SX, Kim A, Madara JC, Zhu PK, Christenson LF, Lutas A, Kalugin PN, Jin Y, Pal A, Tian L, Lowell BB, Andermann ML. Competition between stochastic neuropeptide signals calibrates the rate of satiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.11.548551. [PMID: 37503012 PMCID: PMC10369917 DOI: 10.1101/2023.07.11.548551] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
We investigated how transmission of hunger- and satiety-promoting neuropeptides, NPY and αMSH, is integrated at the level of intracellular signaling to control feeding. Receptors for these peptides use the second messenger cAMP, but the messenger's spatiotemporal dynamics and role in energy balance are controversial. We show that AgRP axon stimulation in the paraventricular hypothalamus evokes probabilistic and spatially restricted NPY release that triggers stochastic cAMP decrements in downstream MC4R-expressing neurons (PVH MC4R ). Meanwhile, POMC axon stimulation triggers stochastic, αMSH-dependent cAMP increments. NPY and αMSH competitively control cAMP, as reflected by hunger-state-dependent differences in the amplitude and persistence of cAMP transients evoked by each peptide. During feeding bouts, elevated αMSH release and suppressed NPY release cooperatively sustain elevated cAMP in PVH MC4R neurons, thereby potentiating feeding-related excitatory inputs and promoting satiation across minutes. Our findings highlight how state-dependent integration of opposing, quantal peptidergic events by a common biochemical target calibrates energy intake.
Collapse
|
59
|
Qian T, Wang H, Wang P, Geng L, Mei L, Osakada T, Wang L, Tang Y, Kania A, Grinevich V, Stoop R, Lin D, Luo M, Li Y. A genetically encoded sensor measures temporal oxytocin release from different neuronal compartments. Nat Biotechnol 2023; 41:944-957. [PMID: 36593404 PMCID: PMC11182738 DOI: 10.1038/s41587-022-01561-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 10/12/2022] [Indexed: 01/03/2023]
Abstract
Oxytocin (OT), a peptide hormone and neuromodulator, is involved in diverse physiological and pathophysiological processes in the central nervous system and the periphery. However, the regulation and functional sequences of spatial OT release in the brain remain poorly understood. We describe a genetically encoded G-protein-coupled receptor activation-based (GRAB) OT sensor called GRABOT1.0. In contrast to previous methods, GRABOT1.0 enables imaging of OT release ex vivo and in vivo with suitable sensitivity, specificity and spatiotemporal resolution. Using this sensor, we visualize stimulation-induced OT release from specific neuronal compartments in mouse brain slices and discover that N-type calcium channels predominantly mediate axonal OT release, whereas L-type calcium channels mediate somatodendritic OT release. We identify differences in the fusion machinery of OT release for axon terminals versus somata and dendrites. Finally, we measure OT dynamics in various brain regions in mice during male courtship behavior. Thus, GRABOT1.0 provides insights into the role of compartmental OT release in physiological and behavioral functions.
Collapse
Affiliation(s)
- Tongrui Qian
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Huan Wang
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Peng Wang
- Medical Center for Human Reproduction, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lan Geng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Long Mei
- Neuroscience Institute, Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - Takuya Osakada
- Neuroscience Institute, Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - Lei Wang
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Peking University, Beijing, China
| | - Yan Tang
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital Center (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Alan Kania
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ron Stoop
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital Center (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Dayu Lin
- Neuroscience Institute, Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - Minmin Luo
- National Institute of Biological Sciences (NIBS), Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), Tsinghua University, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China.
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China.
- Chinese Institute for Brain Research, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
- National Biomedical Imaging Center, Peking University, Beijing, China.
| |
Collapse
|
60
|
Chen S, Rao BY, Herrlinger S, Losonczy A, Paninski L, Varol E. MULTIMODAL MICROSCOPY IMAGE ALIGNMENT USING SPATIAL AND SHAPE INFORMATION AND A BRANCH-AND-BOUND ALGORITHM. PROCEEDINGS OF THE ... IEEE INTERNATIONAL CONFERENCE ON ACOUSTICS, SPEECH, AND SIGNAL PROCESSING. ICASSP (CONFERENCE) 2023; 2023:10.1109/icassp49357.2023.10096185. [PMID: 37388235 PMCID: PMC10308861 DOI: 10.1109/icassp49357.2023.10096185] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
Multimodal microscopy experiments that image the same population of cells under different experimental conditions have become a widely used approach in systems and molecular neuroscience. The main obstacle is to align the different imaging modalities to obtain complementary information about the observed cell population (e.g., gene expression and calcium signal). Traditional image registration methods perform poorly when only a small subset of cells are present in both images, as is common in multimodal experiments. We cast multimodal microscopy alignment as a cell subset matching problem. To solve this non-convex problem, we introduce an efficient and globally optimal branch-and-bound algorithm to find subsets of point clouds that are in rotational alignment with each other. In addition, we use complementary information about cell shape and location to compute the matching likelihood of cell pairs in two imaging modalities to further prune the optimization search tree. Finally, we use the maximal set of cells in rigid rotational alignment to seed image deformation fields to obtain a final registration result. Our framework performs better than the state-of-the-art histology alignment approaches regarding matching quality and is faster than manual alignment, providing a viable solution to improve the throughput of multimodal microscopy experiments.
Collapse
Affiliation(s)
- Shuonan Chen
- Department of System Biology
- Zuckerman Institute
- Columbia University
| | - Bovey Y Rao
- Department of Neurobiology
- Zuckerman Institute
- Columbia University
| | | | - Attila Losonczy
- Department of Neurobiology
- Zuckerman Institute
- Columbia University
| | - Liam Paninski
- Department of Statistics
- Zuckerman Institute
- Columbia University
| | - Erdem Varol
- Department of Statistics
- Department of Computer Science & Engineering
- Zuckerman Institute
- Columbia University
- New York University
| |
Collapse
|
61
|
Park HE, Jo SH, Lee RH, Macks CP, Ku T, Park J, Lee CW, Hur JK, Sohn CH. Spatial Transcriptomics: Technical Aspects of Recent Developments and Their Applications in Neuroscience and Cancer Research. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206939. [PMID: 37026425 PMCID: PMC10238226 DOI: 10.1002/advs.202206939] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 03/10/2023] [Indexed: 06/04/2023]
Abstract
Spatial transcriptomics is a newly emerging field that enables high-throughput investigation of the spatial localization of transcripts and related analyses in various applications for biological systems. By transitioning from conventional biological studies to "in situ" biology, spatial transcriptomics can provide transcriptome-scale spatial information. Currently, the ability to simultaneously characterize gene expression profiles of cells and relevant cellular environment is a paradigm shift for biological studies. In this review, recent progress in spatial transcriptomics and its applications in neuroscience and cancer studies are highlighted. Technical aspects of existing technologies and future directions of new developments (as of March 2023), computational analysis of spatial transcriptome data, application notes in neuroscience and cancer studies, and discussions regarding future directions of spatial multi-omics and their expanding roles in biomedical applications are emphasized.
Collapse
Affiliation(s)
- Han-Eol Park
- Center for Nanomedicine, Institute for Basic Science, Yonsei University, Seoul, 03722, Republic of Korea
- Graduate Program in Nanobiomedical Engineering, Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
- School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Song Hyun Jo
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Rosalind H Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Christian P Macks
- Center for Nanomedicine, Institute for Basic Science, Yonsei University, Seoul, 03722, Republic of Korea
- Graduate Program in Nanobiomedical Engineering, Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Taeyun Ku
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jihwan Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Chung Whan Lee
- Department of Chemistry, Gachon University, Seongnam, Gyeonggi-do, 13120, Republic of Korea
| | - Junho K Hur
- Department of Genetics, College of Medicine, Hanyang University, Seoul, 04763, Republic of Korea
| | - Chang Ho Sohn
- Center for Nanomedicine, Institute for Basic Science, Yonsei University, Seoul, 03722, Republic of Korea
- Graduate Program in Nanobiomedical Engineering, Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| |
Collapse
|
62
|
Wang Y, Ye L. Somatosensory innervation of adipose tissues. Physiol Behav 2023; 265:114174. [PMID: 36965573 PMCID: PMC11537203 DOI: 10.1016/j.physbeh.2023.114174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/27/2023]
Abstract
The increasing prevalence of obesity and type 2 diabetes has led to a greater interest in adipose tissue physiology. Adipose tissue is now understood as an organ with endocrine and thermogenic capacities in addition to its role in fat storage. It plays a critical role in systemic metabolism and energy regulation, and its activity is tightly regulated by the nervous system. Fat is now recognized to receive sympathetic innervation, which transmits information from the brain, as well as sensory innervation, which sends information into the brain. The role of sympathetic innervation in adipose tissue has been extensively studied. However, the extent and the functional significance of sensory innervation have long been unclear. Recent studies have started to reveal that sensory neurons robustly innervate adipose tissue and play an important role in regulating fat activity. This brief review will discuss both historical evidence and recent advances, as well as important remaining questions about the sensory innervation of adipose tissue.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Li Ye
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
63
|
Kin K, Francis-Oliveira J, Kano SI, Niwa M. Adolescent stress impairs postpartum social behavior via anterior insula-prelimbic pathway in mice. Nat Commun 2023; 14:2975. [PMID: 37221211 PMCID: PMC10205810 DOI: 10.1038/s41467-023-38799-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 05/16/2023] [Indexed: 05/25/2023] Open
Abstract
Adolescent stress can be a risk factor for abnormal social behavior in the postpartum period, which critically affects an individual social functioning. Nonetheless, the underlying mechanisms remain unclear. Using a mouse model with optogenetics and in vivo calcium imaging, we found that adolescent psychosocial stress, combined with pregnancy and delivery, caused hypofunction of the glutamatergic pathway from the anterior insula to prelimbic cortex (AI-PrL pathway), which altered PrL neuronal activity, and in turn led to abnormal social behavior. Specifically, the AI-PrL pathway played a crucial role during recognizing the novelty of other mice by modulating "stable neurons" in PrL, which were constantly activated or inhibited by novel mice. We also observed that glucocorticoid receptor signaling in the AI-PrL pathway had a causal role in stress-induced postpartum changes. Our findings provide functional insights into a cortico-cortical pathway underlying adolescent stress-induced postpartum social behavioral deficits.
Collapse
Affiliation(s)
- Kyohei Kin
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, 35233, USA
| | - Jose Francis-Oliveira
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, 35233, USA
| | - Shin-Ichi Kano
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, 35233, USA
- Department of Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, 35233, USA
| | - Minae Niwa
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, 35233, USA.
- Department of Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, 35233, USA.
- Department of Biomedical Engineering, University of Alabama at Birmingham School of Engineering, Birmingham, AL, 22908, USA.
| |
Collapse
|
64
|
Li Q, Lin Z, Liu R, Tang X, Huang J, He Y, Sui X, Tian W, Shen H, Zhou H, Sheng H, Shi H, Xiao L, Wang X, Liu J. Multimodal charting of molecular and functional cell states via in situ electro-sequencing. Cell 2023; 186:2002-2017.e21. [PMID: 37080201 PMCID: PMC11259179 DOI: 10.1016/j.cell.2023.03.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/21/2022] [Accepted: 03/16/2023] [Indexed: 04/22/2023]
Abstract
Paired mapping of single-cell gene expression and electrophysiology is essential to understand gene-to-function relationships in electrogenic tissues. Here, we developed in situ electro-sequencing (electro-seq) that combines flexible bioelectronics with in situ RNA sequencing to stably map millisecond-timescale electrical activity and profile single-cell gene expression from the same cells across intact biological networks, including cardiac and neural patches. When applied to human-induced pluripotent stem-cell-derived cardiomyocyte patches, in situ electro-seq enabled multimodal in situ analysis of cardiomyocyte electrophysiology and gene expression at the cellular level, jointly defining cell states and developmental trajectories. Using machine-learning-based cross-modal analysis, in situ electro-seq identified gene-to-electrophysiology relationships throughout cardiomyocyte development and accurately reconstructed the evolution of gene expression profiles based on long-term stable electrical measurements. In situ electro-seq could be applicable to create spatiotemporal multimodal maps in electrogenic tissues, potentiating the discovery of cell types and gene programs responsible for electrophysiological function and dysfunction.
Collapse
Affiliation(s)
- Qiang Li
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Zuwan Lin
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Ren Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Xin Tang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jiahao Huang
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yichun He
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Xin Sui
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Weiwen Tian
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Hao Shen
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Haowen Zhou
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Hao Sheng
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Hailing Shi
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ling Xiao
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Cardiovascular Disease Initiative and Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Xiao Wang
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Jia Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA.
| |
Collapse
|
65
|
Hostetler RE, Hu H, Agmon A. Genetically Defined Subtypes of Somatostatin-Containing Cortical Interneurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.02.526850. [PMID: 36778499 PMCID: PMC9915678 DOI: 10.1101/2023.02.02.526850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Inhibitory interneurons play a crucial role in proper development and function of the mammalian cerebral cortex. Of the different inhibitory subclasses, dendritic-targeting, somatostatin-containing (SOM) interneurons may be the most diverse. Earlier studies used transgenic mouse lines to identify and characterize subtypes of SOM interneurons by morphological, electrophysiological and neurochemical properties. More recently, large-scale studies classified SOM interneurons into 13 morpho-electro-transcriptomic (MET) types. It remains unclear, however, how these various classification schemes relate to each other, and experimental access to MET types has been limited by the scarcity of type-specific mouse driver lines. To begin to address these issues we crossed Flp and Cre driver mouse lines and a dual-color combinatorial reporter, allowing experimental access to genetically defined SOM subsets. Brains from adult mice of both sexes were retrogradely dye-labeled from the pial surface to identify layer 1-projecting neurons, and immunostained against several marker proteins, allowing correlation of genetic label, axonal target and marker protein expression in the same neurons. Using whole-cell recordings ex-vivo, we compared electrophysiological properties between intersectional and transgenic SOM subsets. We identified two layer 1-targeting intersectional subsets with non-overlapping marker protein expression and electrophysiological properties which, together with a previously characterized layer 4-targeting subtype, account for about half of all layer 5 SOM cells and >40% of all SOM cells, and appear to map onto 5 of the 13 MET types. Genetic access to these subtypes will allow researchers to determine their synaptic inputs and outputs and uncover their roles in cortical computations and animal behavior. SIGNIFICANCE STATEMENT Inhibitory neurons are critically important for proper development and function of the cerebral cortex. Although a minority population, they are highly diverse, which poses a major challenge to investigating their contributions to cortical computations and animal and human behavior. As a step towards understanding this diversity we crossed genetically modified mouse lines to allow detailed examination of genetically-defined groups of the most diverse inhibitory subtype, somatostatin-containing interneurons. We identified and characterized three somatostatin subtypes in the deep cortical layers with distinct combinations of anatomical, neurochemical and electrophysiological properties. Future studies could now use these genetic tools to examine how these different subtypes are integrated into the cortical circuit and what roles they play during sensory, cognitive or motor behavior.
Collapse
Affiliation(s)
- Rachel E Hostetler
- Dept. of Neuroscience, West Virginia University School of Medicine, WV Rockefeller Neuroscience Institute, Morgantown, WV 26506, USA
| | - Hang Hu
- Dept. of Neuroscience, West Virginia University School of Medicine, WV Rockefeller Neuroscience Institute, Morgantown, WV 26506, USA
| | - Ariel Agmon
- Dept. of Neuroscience, West Virginia University School of Medicine, WV Rockefeller Neuroscience Institute, Morgantown, WV 26506, USA
| |
Collapse
|
66
|
Bárez-López S, Mecawi AS, Bryan N, Pauža AG, Duque VJ, Gillard BT, Murphy D, Greenwood MP. Translational and post-translational dynamics in a model peptidergic system. Mol Cell Proteomics 2023; 22:100544. [PMID: 37030596 DOI: 10.1016/j.mcpro.2023.100544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 03/30/2023] [Accepted: 04/04/2023] [Indexed: 04/10/2023] Open
Abstract
The cell bodies of hypothalamic magnocellular neurones are densely packed in the hypothalamic supraoptic nucleus whereas their axons project to the anatomically discrete posterior pituitary gland. We have taken advantage of this unique anatomical structure to establish proteome and phosphoproteome dynamics in neuronal cell bodies and axonal terminals in response to physiological stimulation. We have found that proteome and phosphoproteome responses to neuronal stimulation are very different between somatic and axonal neuronal compartments, indicating the need of each cell domain to differentially adapt. In particular, changes in the phosphoproteome in the cell body are involved in the reorganisation of the cytoskeleton and in axonal terminals the regulation of synaptic and secretory processes. We have identified that prohormone precursors including vasopressin and oxytocin are phosphorylated in axonal terminals and are hyperphosphorylated following stimulation. By multi-omic integration of transcriptome and proteomic data we identify changes to proteins present in afferent inputs to this nucleus.
Collapse
Affiliation(s)
- Soledad Bárez-López
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Bristol, United Kingdom
| | - André S Mecawi
- Laboratory of Molecular Neuroendocrinology, Department of Biophysics, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Natasha Bryan
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Bristol, United Kingdom
| | - Audrys G Pauža
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Bristol, United Kingdom
| | - Victor J Duque
- Laboratory of Molecular Neuroendocrinology, Department of Biophysics, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Benjamin T Gillard
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Bristol, United Kingdom
| | - David Murphy
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Bristol, United Kingdom.
| | - Michael P Greenwood
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Bristol, United Kingdom.
| |
Collapse
|
67
|
Kin K, Francis-Oliveira J, Kano SI, Niwa M. Adolescent stress impairs postpartum social behavior via anterior insula-prelimbic pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.03.522598. [PMID: 36711960 PMCID: PMC9881883 DOI: 10.1101/2023.01.03.522598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Adolescent stress can be a risk factor for abnormal social behavior in the postpartum period, which critically affects the safety of mothers and children. Nonetheless, the underlying mechanisms remain unclear. Using a newly established mouse model with optogenetics and in vivo calcium imaging, we found that adolescent psychosocial stress, combined with pregnancy and delivery, caused hypofunction of the glutamatergic pathway from the anterior insula to prelimbic cortex (AI-PrL pathway), which altered PrL neuronal activity, and in turn led to abnormal social behavior. Specifically, the AI-PrL pathway played a crucial role during recognizing the novelty of other mice by modulating ″stable neurons″ in PrL, which were constantly activated or inhibited by novel mice. We also observed that glucocorticoid receptor signaling in the AI-PrL pathway had a causal role in stress-induced postpartum changes. Our findings provide novel and functional insights into a cortico-cortical pathway underlying adolescent stress-induced postpartum social behavioral deficits.
Collapse
Affiliation(s)
- Kyohei Kin
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Jose Francis-Oliveira
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Shin-ichi Kano
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
- Department of Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Minae Niwa
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
- Department of Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham School of Engineering, Birmingham, AL 22908, USA
| |
Collapse
|
68
|
Zhang Y, Petukhov V, Biederstedt E, Que R, Zhang K, Kharchenko PV. Gene panel selection for targeted spatial transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.03.527053. [PMID: 36993340 PMCID: PMC10054990 DOI: 10.1101/2023.02.03.527053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Targeted spatial transcriptomics hold particular promise in analysis of complex tissues. Most such methods, however, measure only a limited panel of transcripts, which need to be selected in advance to inform on the cell types or processes being studied. A limitation of existing gene selection methods is that they rely on scRNA-seq data, ignoring platform effects between technologies. Here we describe gpsFISH, a computational method to perform gene selection through optimizing detection of known cell types. By modeling and adjusting for platform effects, gpsFISH outperforms other methods. Furthermore, gpsFISH can incorporate cell type hierarchies and custom gene preferences to accommodate diverse design requirements.
Collapse
Affiliation(s)
- Yida Zhang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
| | - Viktor Petukhov
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Evan Biederstedt
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Richard Que
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Kun Zhang
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- San Diego Institute of Science, Altos Labs, San Diego, CA, USA
| | - Peter V. Kharchenko
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- San Diego Institute of Science, Altos Labs, San Diego, CA, USA
| |
Collapse
|
69
|
Zhang M, Pan X, Jung W, Halpern A, Eichhorn SW, Lei Z, Cohen L, Smith KA, Tasic B, Yao Z, Zeng H, Zhuang X. A molecularly defined and spatially resolved cell atlas of the whole mouse brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.06.531348. [PMID: 36945367 PMCID: PMC10028822 DOI: 10.1101/2023.03.06.531348] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
In mammalian brains, tens of millions to billions of cells form complex interaction networks to enable a wide range of functions. The enormous diversity and intricate organization of cells in the brain have so far hindered our understanding of the molecular and cellular basis of its functions. Recent advances in spatially resolved single-cell transcriptomics have allowed systematic mapping of the spatial organization of molecularly defined cell types in complex tissues1-3. However, these approaches have only been applied to a few brain regions1-11 and a comprehensive cell atlas of the whole brain is still missing. Here, we imaged a panel of >1,100 genes in ~8 million cells across the entire adult mouse brain using multiplexed error-robust fluorescence in situ hybridization (MERFISH)12 and performed spatially resolved, single-cell expression profiling at the whole-transcriptome scale by integrating MERFISH and single-cell RNA-sequencing (scRNA-seq) data. Using this approach, we generated a comprehensive cell atlas of >5,000 transcriptionally distinct cell clusters, belonging to ~300 major cell types, in the whole mouse brain with high molecular and spatial resolution. Registration of the MERFISH images to the common coordinate framework (CCF) of the mouse brain further allowed systematic quantifications of the cell composition and organization in individual brain regions defined in the CCF. We further identified spatial modules characterized by distinct cell-type compositions and spatial gradients featuring gradual changes in the gene-expression profiles of cells. Finally, this high-resolution spatial map of cells, with a transcriptome-wide expression profile associated with each cell, allowed us to infer cell-type-specific interactions between several hundred pairs of molecularly defined cell types and predict potential molecular (ligand-receptor) basis and functional implications of these cell-cell interactions. These results provide rich insights into the molecular and cellular architecture of the brain and a valuable resource for future functional investigations of neural circuits and their dysfunction in diseases.
Collapse
Affiliation(s)
- Meng Zhang
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
- These authors contributed equally
| | - Xingjie Pan
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
- These authors contributed equally
| | - Won Jung
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
- These authors contributed equally
| | - Aaron Halpern
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
| | - Stephen W. Eichhorn
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
| | - Zhiyun Lei
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
| | - Limor Cohen
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
| | | | - Bosiljka Tasic
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Xiaowei Zhuang
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
70
|
Petzold A, van den Munkhof HE, Figge-Schlensok R, Korotkova T. Complementary lateral hypothalamic populations resist hunger pressure to balance nutritional and social needs. Cell Metab 2023; 35:456-471.e6. [PMID: 36827985 PMCID: PMC10028225 DOI: 10.1016/j.cmet.2023.02.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 05/03/2022] [Accepted: 02/08/2023] [Indexed: 02/25/2023]
Abstract
Animals continuously weigh hunger and thirst against competing needs, such as social contact and mating, according to state and opportunity. Yet neuronal mechanisms of sensing and ranking nutritional needs remain poorly understood. Here, combining calcium imaging in freely behaving mice, optogenetics, and chemogenetics, we show that two neuronal populations of the lateral hypothalamus (LH) guide increasingly hungry animals through behavioral choices between nutritional and social rewards. While increased food consumption was marked by increasing inhibition of a leptin receptor-expressing (LepRLH) subpopulation at a fast timescale, LepRLH neurons limited feeding or drinking and promoted social interaction despite hunger or thirst. Conversely, neurotensin-expressing LH neurons preferentially encoded water despite hunger pressure and promoted water seeking, while relegating social needs. Thus, hunger and thirst gate both LH populations in a complementary manner to enable the flexible fulfillment of multiple essential needs.
Collapse
Affiliation(s)
- Anne Petzold
- Institute for Systems Physiology, Faculty of Medicine, University of Cologne and University Clinic Cologne, Cologne 50931, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Hanna Elin van den Munkhof
- Institute for Systems Physiology, Faculty of Medicine, University of Cologne and University Clinic Cologne, Cologne 50931, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Rebecca Figge-Schlensok
- Institute for Systems Physiology, Faculty of Medicine, University of Cologne and University Clinic Cologne, Cologne 50931, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Tatiana Korotkova
- Institute for Systems Physiology, Faculty of Medicine, University of Cologne and University Clinic Cologne, Cologne 50931, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne 50931, Germany.
| |
Collapse
|
71
|
Integrated cardio-behavioral responses to threat define defensive states. Nat Neurosci 2023; 26:447-457. [PMID: 36759559 PMCID: PMC9991919 DOI: 10.1038/s41593-022-01252-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 12/21/2022] [Indexed: 02/11/2023]
Abstract
Fear and anxiety are brain states that evolved to mediate defensive responses to threats. The defense reaction includes multiple interacting behavioral, autonomic and endocrine adjustments, but their integrative nature is poorly understood. In particular, although threat has been associated with various cardiac changes, there is no clear consensus regarding the relevance of these changes for the integrated defense reaction. Here we identify rapid microstates that are associated with specific behaviors and heart rate dynamics, which are affected by long-lasting macrostates and reflect context-dependent threat levels. In addition, we demonstrate that one of the most commonly used defensive behavioral responses-freezing as measured by immobility-is part of an integrated cardio-behavioral microstate mediated by Chx10+ neurons in the periaqueductal gray. Our framework for systematic integration of cardiac and behavioral readouts presents the basis for a better understanding of complex neural defensive states and their associated systemic functions.
Collapse
|
72
|
Ledford H. Sex, food or water? How mice decide. Nature 2023:10.1038/d41586-023-00521-3. [PMID: 36823270 DOI: 10.1038/d41586-023-00521-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
|
73
|
Shainer I, Kuehn E, Laurell E, Al Kassar M, Mokayes N, Sherman S, Larsch J, Kunst M, Baier H. A single-cell resolution gene expression atlas of the larval zebrafish brain. SCIENCE ADVANCES 2023; 9:eade9909. [PMID: 36812331 PMCID: PMC9946346 DOI: 10.1126/sciadv.ade9909] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/22/2022] [Indexed: 06/18/2023]
Abstract
The advent of multimodal brain atlases promises to accelerate progress in neuroscience by allowing in silico queries of neuron morphology, connectivity, and gene expression. We used multiplexed fluorescent in situ RNA hybridization chain reaction (HCR) technology to generate expression maps across the larval zebrafish brain for a growing set of marker genes. The data were registered to the Max Planck Zebrafish Brain (mapzebrain) atlas, thus allowing covisualization of gene expression, single-neuron tracings, and expertly curated anatomical segmentations. Using post hoc HCR labeling of the immediate early gene cfos, we mapped responses to prey stimuli and food ingestion across the brain of freely swimming larvae. This unbiased approach revealed, in addition to previously described visual and motor areas, a cluster of neurons in the secondary gustatory nucleus, which express the marker calb2a, as well as a specific neuropeptide Y receptor, and project to the hypothalamus. This discovery exemplifies the power of this new atlas resource for zebrafish neurobiology.
Collapse
|
74
|
Meijer OC, Buurstede JC, Viho EMG, Amaya JM, Koning ASCAM, van der Meulen M, van Weert LTCM, Paul SN, Kroon J, Koorneef LL. Transcriptional glucocorticoid effects in the brain: Finding the relevant target genes. J Neuroendocrinol 2023; 35:e13213. [PMID: 36426812 DOI: 10.1111/jne.13213] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 10/25/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022]
Abstract
Glucocorticoids are powerful modulators of brain function. They act via mineralocorticoid and glucocorticoid receptors (MR and GR). These are best understood as transcription factors. Although many glucocorticoid effects depend on the modulation of gene transcription, it is a major challenge to link gene expression to function given the large-scale, apparently pleiotropic genomic responses. The extensive sets of MR and GR target genes are highly specific per cell type, and the brain contains many different (neuronal and non-neuronal) cell types. Next to the set "trait" of cellular context, the "state" of other active signaling pathways will affect MR and GR transcriptional activity. Here, we discuss receptor specificity and contextual factors that determine the transcriptional outcome of MR/GR signaling, experimental possibilities offered by single-cell transcriptomics approaches, and reflect on how to make sense of lists of target genes in relation to understanding the functional effects of steroid receptor activation.
Collapse
Affiliation(s)
- Onno C Meijer
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jacobus C Buurstede
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Eva M G Viho
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jorge Miguel Amaya
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Anne-Sophie C A M Koning
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Merel van der Meulen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Lisa T C M van Weert
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Susana N Paul
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Kroon
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Lisa L Koorneef
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
75
|
Yao P, Liu R, Broginni T, Thunemann M, Kleinfeld D. Guide to the construction and use of an adaptive optics two-photon microscope with direct wavefront sensing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.24.525307. [PMID: 36747816 PMCID: PMC9900836 DOI: 10.1101/2023.01.24.525307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Two-photon microscopy, combined with appropriate optical labeling, has enabled the study of structure and function throughout nervous systems. This methodology enables, for example, the measurement and tracking of sub-micrometer structures within brain cells, the spatio-temporal mapping of spikes in individual neurons, and the spatio-temporal mapping of transmitter release in individual synapses. Yet the spatial resolution of two-photon microscopy rapidly degrades as imaging is attempted at depths more than a few scattering lengths into tissue, i.e., below the superficial layers that constitute the top 300 to 400 µm of neocortex. To obviate this limitation, we measure the wavefront at the focus of the excitation beam and utilize adaptive optics that alters the incident wavefront to achieve an improved focal volume. We describe the constructions, calibration, and operation of a two-photon microscopy that incorporates adaptive optics to restore diffraction-limited resolution throughout the nearly 900 µm depth of mouse cortex. Our realization utilizes a guide star formed by excitation of red-shifted dye within the blood serum to directly measure the wavefront. We incorporate predominantly commercial optical, optomechanical, mechanical, and electronic components; computer aided design models of the exceptional custom components are supplied. The design is modular and allows for expanded imaging and optical excitation capabilities. We demonstrate our methodology in mouse neocortex by imaging the morphology of somatostatin-expressing neurons at 700 µm beneath the pia, calcium dynamics of layer 5b projection neurons, and glutamate transmission to L4 neurons.
Collapse
|
76
|
Bárez-López S, Scanlon L, Murphy D, Greenwood MP. Imaging the Hypothalamo-Neurohypophysial System. Neuroendocrinology 2023; 113:168-178. [PMID: 34438401 DOI: 10.1159/000519233] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/23/2021] [Indexed: 11/19/2022]
Abstract
The hypothalamo-neurohypophysial system (HNS) is a brain peptidergic neurosecretory apparatus which is composed of arginine vasopressin (AVP) and oxytocin (OXT) magnocellular neurones and their neuronal processes in the posterior pituitary (PP). In response to specific stimuli, AVP and OXT are secreted into the systemic circulation at the neurovascular interface of the PP, where they act as hormones, but they can also behave as neurotransmitters when released at the somatodendritic compartment or by axon collaterals to other brain regions. Because these peptides are crucial for several physiological processes, including fluid homoeostasis and reproduction, it is of great importance to map the HNS connectome in its entirety in order to understand its functions. In recent years, advances in imaging technologies have provided considerable new information about the HNS. These approaches include the use of reporter proteins under the control of specific promoters, viral tracers, brain-clearing methods, genetically encoded indicators, sniffer cells, mass spectrometry imaging, and spatially resolved transcriptomics. In this review, we illustrate how these latest approaches have enhanced our understanding of the structure and function of the HNS and how they might contribute further in the coming years.
Collapse
Affiliation(s)
- Soledad Bárez-López
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Liam Scanlon
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - David Murphy
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Michael Paul Greenwood
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
77
|
Deng Y, Dickey JE, Saito K, Deng G, Singh U, Jiang J, Toth BA, Zhu Z, Zingman LV, Resch JM, Grobe JL, Cui H. Elucidating the role of Rgs2 expression in the PVN for metabolic homeostasis in mice. Mol Metab 2022; 66:101622. [PMID: 36307046 PMCID: PMC9638802 DOI: 10.1016/j.molmet.2022.101622] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/09/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE RGS2 is a GTPase activating protein that modulates GPCR-Gα signaling and mice lacking RGS2 globally exhibit metabolic alterations. While RGS2 is known to be broadly expressed throughout the body including the brain, the relative contribution of brain RGS2 to metabolic homeostasis remains unknown. The purpose of this study was to characterize RGS2 expression in the paraventricular nucleus of hypothalamus (PVN) and test its role in metabolic homeostasis. METHODS We used a combination of RNAscope in situ hybridization (ISH), immunohistochemistry, and bioinformatic analyses to characterize the pattern of Rgs2 expression in the PVN. We then created mice lacking Rgs2 either prenatally or postnatally in the PVN and evaluated their metabolic consequences. RESULTS RNAscope ISH analysis revealed a broad but regionally enriched Rgs2 mRNA expression throughout the mouse brain, with the highest expression being observed in the PVN along with several other brain regions, such as the arcuate nucleus of hypothalamus and the dorsal raphe nucleus. Within the PVN, we found that Rgs2 is specifically enriched in CRH+ endocrine neurons and is further increased by calorie restriction. Functionally, although Sim1-Cre-mediated prenatal deletion of Rgs2 in PVN neurons had no major effects on metabolic homeostasis, AAV-mediated adult deletion of Rgs2 in the PVN led to significantly increased food intake, body weight (both fat and fat-free masses), body length, and blood glucose levels in both male and female mice. Strikingly, we found that prolonged postnatal loss of Rgs2 leads to neuronal cell death in the PVN, while rapid body weight gain in the early phase of viral-mediated PVN Rgs2 deletion is independent of PVN neuronal loss. CONCLUSIONS Our results provide the first evidence to show that PVN Rgs2 expression is not only sensitive to metabolic challenge but also critically required for PVN endocrine neurons to function and maintain metabolic homeostasis.
Collapse
Affiliation(s)
- Yue Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Jacob E Dickey
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Kenji Saito
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Guorui Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Uday Singh
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Jingwei Jiang
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Brandon A Toth
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Zhiyong Zhu
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Leonid V Zingman
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, United States; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Jon M Resch
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States; F.O.E. Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Justin L Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Huxing Cui
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States; F.O.E. Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, United States.
| |
Collapse
|
78
|
Watts AG. Paraventricular nucleus-Medullary interactions: How they help enable endocrine responses to metabolic stress. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2022; 27:100401. [PMID: 39575062 PMCID: PMC11580161 DOI: 10.1016/j.coemr.2022.100401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2024]
Abstract
The paraventricular hypothalamic nucleus (PVH) organizes neuroendocrine and autonomic responses to rapidly and slowly developing metabolic stressors that limit their impact on energy balance. The PVH together with the lateral hypothalamic area, and the arcuate and dorsomedial nuclei form a network that is defined by its inputs from medullary catecholamine neurons. These medullary neurons convey important glycemia and glucocorticoid feedback information that is integrated by the PVH and the rest of this network to control a variety of responses to metabolic stressors that have rapid (hypoglycemia) or slow onsets (eating a high calorie diet). This review focuses on how the responses to these two challenges are enabled by these catecholamine neurons, and the integrative nature of the network into which they project.
Collapse
Affiliation(s)
- Alan G Watts
- The Department of Biological Sciences, USC Dornsife College of Letters, Arts & Sciences, University of Southern California, Los Angeles, USA
| |
Collapse
|
79
|
Luxem K, Mocellin P, Fuhrmann F, Kürsch J, Miller SR, Palop JJ, Remy S, Bauer P. Identifying behavioral structure from deep variational embeddings of animal motion. Commun Biol 2022; 5:1267. [PMID: 36400882 PMCID: PMC9674640 DOI: 10.1038/s42003-022-04080-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 10/06/2022] [Indexed: 11/19/2022] Open
Abstract
Quantification and detection of the hierarchical organization of behavior is a major challenge in neuroscience. Recent advances in markerless pose estimation enable the visualization of high-dimensional spatiotemporal behavioral dynamics of animal motion. However, robust and reliable technical approaches are needed to uncover underlying structure in these data and to segment behavior into discrete hierarchically organized motifs. Here, we present an unsupervised probabilistic deep learning framework that identifies behavioral structure from deep variational embeddings of animal motion (VAME). By using a mouse model of beta amyloidosis as a use case, we show that VAME not only identifies discrete behavioral motifs, but also captures a hierarchical representation of the motif's usage. The approach allows for the grouping of motifs into communities and the detection of differences in community-specific motif usage of individual mouse cohorts that were undetectable by human visual observation. Thus, we present a robust approach for the segmentation of animal motion that is applicable to a wide range of experimental setups, models and conditions without requiring supervised or a-priori human interference.
Collapse
Affiliation(s)
- Kevin Luxem
- Leibniz Institute for Neurobiology (LIN), Department of Cellular Neuroscience, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Petra Mocellin
- Leibniz Institute for Neurobiology (LIN), Department of Cellular Neuroscience, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Falko Fuhrmann
- Leibniz Institute for Neurobiology (LIN), Department of Cellular Neuroscience, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Johannes Kürsch
- Leibniz Institute for Neurobiology (LIN), Department of Cellular Neuroscience, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Stephanie R Miller
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Jorge J Palop
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Stefan Remy
- Leibniz Institute for Neurobiology (LIN), Department of Cellular Neuroscience, Magdeburg, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.
- German Center for Mental Health (DZPG), Magdeburg, Germany.
| | - Pavol Bauer
- Leibniz Institute for Neurobiology (LIN), Department of Cellular Neuroscience, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| |
Collapse
|
80
|
Machado TA, Kauvar IV, Deisseroth K. Multiregion neuronal activity: the forest and the trees. Nat Rev Neurosci 2022; 23:683-704. [PMID: 36192596 PMCID: PMC10327445 DOI: 10.1038/s41583-022-00634-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2022] [Indexed: 12/12/2022]
Abstract
The past decade has witnessed remarkable advances in the simultaneous measurement of neuronal activity across many brain regions, enabling fundamentally new explorations of the brain-spanning cellular dynamics that underlie sensation, cognition and action. These recently developed multiregion recording techniques have provided many experimental opportunities, but thoughtful consideration of methodological trade-offs is necessary, especially regarding field of view, temporal acquisition rate and ability to guarantee cellular resolution. When applied in concert with modern optogenetic and computational tools, multiregion recording has already made possible fundamental biological discoveries - in part via the unprecedented ability to perform unbiased neural activity screens for principles of brain function, spanning dozens of brain areas and from local to global scales.
Collapse
Affiliation(s)
- Timothy A Machado
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Isaac V Kauvar
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
| |
Collapse
|
81
|
Jiang S, Chen L, Huang ZL, Chen CR. Role of the paraventricular nucleus of the hypothalamus in sleep–wake regulation. BRAIN SCIENCE ADVANCES 2022. [DOI: 10.26599/bsa.2022.9050017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The paraventricular nucleus of the hypothalamus (PVH) acts as a cohesive functional unit that regulates neuroendocrine and autonomic function, complex behavior, and negative emotions after stress. However, how the PVH integrates arousal with these biological functions has only recently been explored. Clinical reports, combined with neurotoxic lesioning, immunochemistry, neuronal activity recordings, and the polysomnographic analyses of genetically modified animals, have revealed that the PVH is important for the control of wakefulness. Here, we review emerging anatomical and neural mechanisms for sleep–wake regulation in the PVH to support its essential role in the promotion and maintenance of wakefulness.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College Fudan University, Shanghai 200030, China
| | - Lu Chen
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College Fudan University, Shanghai 200030, China
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College Fudan University, Shanghai 200030, China
| | - Chang-Rui Chen
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College Fudan University, Shanghai 200030, China
| |
Collapse
|
82
|
Flavell SW, Gogolla N, Lovett-Barron M, Zelikowsky M. The emergence and influence of internal states. Neuron 2022; 110:2545-2570. [PMID: 35643077 PMCID: PMC9391310 DOI: 10.1016/j.neuron.2022.04.030] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 02/11/2022] [Accepted: 04/27/2022] [Indexed: 01/09/2023]
Abstract
Animal behavior is shaped by a variety of "internal states"-partially hidden variables that profoundly shape perception, cognition, and action. The neural basis of internal states, such as fear, arousal, hunger, motivation, aggression, and many others, is a prominent focus of research efforts across animal phyla. Internal states can be inferred from changes in behavior, physiology, and neural dynamics and are characterized by properties such as pleiotropy, persistence, scalability, generalizability, and valence. To date, it remains unclear how internal states and their properties are generated by nervous systems. Here, we review recent progress, which has been driven by advances in behavioral quantification, cellular manipulations, and neural population recordings. We synthesize research implicating defined subsets of state-inducing cell types, widespread changes in neural activity, and neuromodulation in the formation and updating of internal states. In addition to highlighting the significance of these findings, our review advocates for new approaches to clarify the underpinnings of internal brain states across the animal kingdom.
Collapse
Affiliation(s)
- Steven W Flavell
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Nadine Gogolla
- Emotion Research Department, Max Planck Institute of Psychiatry, 80804 Munich, Germany; Circuits for Emotion Research Group, Max Planck Institute of Neurobiology, 82152 Martinsried, Germany.
| | - Matthew Lovett-Barron
- Division of Biological Sciences-Neurobiology Section, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Moriel Zelikowsky
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
83
|
Benevento M, Hökfelt T, Harkany T. Ontogenetic rules for the molecular diversification of hypothalamic neurons. Nat Rev Neurosci 2022; 23:611-627. [PMID: 35906427 DOI: 10.1038/s41583-022-00615-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2022] [Indexed: 11/09/2022]
Abstract
The hypothalamus is an evolutionarily conserved endocrine interface that, among other roles, links central homeostatic control to adaptive bodily responses by releasing hormones and neuropeptides from its many neuronal subtypes. In its preoptic, anterior, tuberal and mammillary subdivisions, a kaleidoscope of magnocellular and parvocellular neuroendocrine command neurons, local-circuit neurons, and neurons that project to extrahypothalamic areas are intermingled in partially overlapping patches of nuclei. Molecular fingerprinting has produced data of unprecedented mass and depth to distinguish and even to predict the synaptic and endocrine competences, connectivity and stimulus selectivity of many neuronal modalities. These new insights support eminent studies from the past century but challenge others on the molecular rules that shape the developmental segregation of hypothalamic neuronal subtypes and their use of morphogenic cues for terminal differentiation. Here, we integrate single-cell RNA sequencing studies with those of mouse genetics and endocrinology to describe key stages of hypothalamus development, including local neurogenesis, the direct terminal differentiation of glutamatergic neurons, transition cascades for GABAergic and GABAergic cell-derived dopamine cells, waves of local neuronal migration, and sequential enrichment in neuropeptides and hormones. We particularly emphasize how transcription factors determine neuronal identity and, consequently, circuit architecture, and whether their deviations triggered by environmental factors and hormones provoke neuroendocrine illnesses.
Collapse
Affiliation(s)
- Marco Benevento
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Tomas Hökfelt
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, Solna, Sweden
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria. .,Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
84
|
Islam MT, Rumpf F, Tsuno Y, Kodani S, Sakurai T, Matsui A, Maejima T, Mieda M. Vasopressin neurons in the paraventricular hypothalamus promote wakefulness via lateral hypothalamic orexin neurons. Curr Biol 2022; 32:3871-3885.e4. [PMID: 35907397 DOI: 10.1016/j.cub.2022.07.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 06/11/2022] [Accepted: 07/08/2022] [Indexed: 01/25/2023]
Abstract
The sleep-wakefulness cycle is regulated by complicated neural networks that include many different populations of neurons throughout the brain. Arginine vasopressin neurons in the paraventricular nucleus of the hypothalamus (PVHAVP) regulate various physiological events and behaviors, such as body-fluid homeostasis, blood pressure, stress response, social interaction, and feeding. Changes in arousal level often accompany these PVHAVP-mediated adaptive responses. However, the contribution of PVHAVP neurons to sleep-wakefulness regulation has remained unknown. Here, we report the involvement of PVHAVP neurons in arousal promotion. Optogenetic stimulation of PVHAVP neurons rapidly induced transitions to wakefulness from both NREM and REM sleep. This arousal effect was dependent on AVP expression in these neurons. Similarly, chemogenetic activation of PVHAVP neurons increased wakefulness and reduced NREM and REM sleep, whereas chemogenetic inhibition of these neurons significantly reduced wakefulness and increased NREM sleep. We observed dense projections of PVHAVP neurons in the lateral hypothalamus with potential connections to orexin/hypocretin (LHOrx) neurons. Optogenetic stimulation of PVHAVP neuronal fibers in the LH immediately induced wakefulness, whereas blocking orexin receptors attenuated the arousal effect of PVHAVP neuronal activation drastically. Monosynaptic rabies-virus tracing revealed that PVHAVP neurons receive inputs from multiple brain regions involved in sleep-wakefulness regulation, as well as those involved in stress response and energy metabolism. Moreover, PVHAVP neurons mediated the arousal induced by novelty stress and a melanocortin receptor agonist melanotan-II. Thus, our data suggested that PVHAVP neurons promote wakefulness via LHOrx neurons in the basal sleep-wakefulness and some stressful conditions.
Collapse
Affiliation(s)
- Md Tarikul Islam
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Florian Rumpf
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan; Graduate School of Life Sciences, University of Würzburg, Beatrice-Edgell-Weg 21, 97074 Würzburg, Germany
| | - Yusuke Tsuno
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Shota Kodani
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Takeshi Sakurai
- Faculty of Medicine/WPI-IIIS, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Ayako Matsui
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Takashi Maejima
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Michihiro Mieda
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan.
| |
Collapse
|
85
|
Zeng H. What is a cell type and how to define it? Cell 2022; 185:2739-2755. [PMID: 35868277 DOI: 10.1016/j.cell.2022.06.031] [Citation(s) in RCA: 202] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 12/20/2022]
Abstract
Cell types are the basic functional units of an organism. Cell types exhibit diverse phenotypic properties at multiple levels, making them challenging to define, categorize, and understand. This review provides an overview of the basic principles of cell types rooted in evolution and development and discusses approaches to characterize and classify cell types and investigate how they contribute to the organism's function, using the mammalian brain as a primary example. I propose a roadmap toward a conceptual framework and knowledge base of cell types that will enable a deeper understanding of the dynamic changes of cellular function under healthy and diseased conditions.
Collapse
Affiliation(s)
- Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA 98109, USA.
| |
Collapse
|
86
|
Zeng H, de Vries SEJ. A gene-expression axis defines neuron behaviour. Nature 2022; 607:243-244. [DOI: 10.1038/d41586-022-01640-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
87
|
Bugeon S, Duffield J, Dipoppa M, Ritoux A, Prankerd I, Nicoloutsopoulos D, Orme D, Shinn M, Peng H, Forrest H, Viduolyte A, Reddy CB, Isogai Y, Carandini M, Harris KD. A transcriptomic axis predicts state modulation of cortical interneurons. Nature 2022; 607:330-338. [PMID: 35794483 PMCID: PMC9279161 DOI: 10.1038/s41586-022-04915-7] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 05/27/2022] [Indexed: 12/14/2022]
Abstract
Transcriptomics has revealed that cortical inhibitory neurons exhibit a great diversity of fine molecular subtypes1-6, but it is not known whether these subtypes have correspondingly diverse patterns of activity in the living brain. Here we show that inhibitory subtypes in primary visual cortex (V1) have diverse correlates with brain state, which are organized by a single factor: position along the main axis of transcriptomic variation. We combined in vivo two-photon calcium imaging of mouse V1 with a transcriptomic method to identify mRNA for 72 selected genes in ex vivo slices. We classified inhibitory neurons imaged in layers 1-3 into a three-level hierarchy of 5 subclasses, 11 types and 35 subtypes using previously defined transcriptomic clusters3. Responses to visual stimuli differed significantly only between subclasses, with cells in the Sncg subclass uniformly suppressed, and cells in the other subclasses predominantly excited. Modulation by brain state differed at all hierarchical levels but could be largely predicted from the first transcriptomic principal component, which also predicted correlations with simultaneously recorded cells. Inhibitory subtypes that fired more in resting, oscillatory brain states had a smaller fraction of their axonal projections in layer 1, narrower spikes, lower input resistance and weaker adaptation as determined in vitro7, and expressed more inhibitory cholinergic receptors. Subtypes that fired more during arousal had the opposite properties. Thus, a simple principle may largely explain how diverse inhibitory V1 subtypes shape state-dependent cortical processing.
Collapse
Affiliation(s)
- Stéphane Bugeon
- UCL Queen Square Institute of Neurology, University College London, London, UK.
| | - Joshua Duffield
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Mario Dipoppa
- UCL Queen Square Institute of Neurology, University College London, London, UK
- Center for Theoretical Neuroscience, Columbia University, New York, NY, USA
| | - Anne Ritoux
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Isabelle Prankerd
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | - David Orme
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Maxwell Shinn
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Han Peng
- Department of Physics, University of Oxford, Oxford, UK
| | - Hamish Forrest
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Aiste Viduolyte
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Charu Bai Reddy
- UCL Queen Square Institute of Neurology, University College London, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Yoh Isogai
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, UK
| | - Matteo Carandini
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Kenneth D Harris
- UCL Queen Square Institute of Neurology, University College London, London, UK.
| |
Collapse
|
88
|
Alcantara IC, Tapia APM, Aponte Y, Krashes MJ. Acts of appetite: neural circuits governing the appetitive, consummatory, and terminating phases of feeding. Nat Metab 2022; 4:836-847. [PMID: 35879462 PMCID: PMC10852214 DOI: 10.1038/s42255-022-00611-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 06/16/2022] [Indexed: 12/11/2022]
Abstract
The overconsumption of highly caloric and palatable foods has caused a surge in obesity rates in the past half century, thereby posing a healthcare challenge due to the array of comorbidities linked to heightened body fat accrual. Developing treatments to manage body weight requires a grasp of the neurobiological basis of appetite. In this Review, we discuss advances in neuroscience that have identified brain regions and neural circuits that coordinate distinct phases of eating: food procurement, food consumption, and meal termination. While pioneering work identified several hypothalamic nuclei to be involved in feeding, more recent studies have explored how neuronal populations beyond the hypothalamus, such as the mesolimbic pathway and nodes in the hindbrain, interconnect to modulate appetite. We also examine how long-term exposure to a calorically dense diet rewires feeding circuits and alters the response of motivational systems to food. Understanding how the nervous system regulates eating behaviour will bolster the development of medical strategies that will help individuals to maintain a healthy body weight.
Collapse
Affiliation(s)
- Ivan C Alcantara
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
- Department of Neuroscience, Brown University, Providence, RI, USA
| | | | - Yeka Aponte
- National Institute on Drug Abuse (NIDA), National Institutes of Health, Baltimore, MD, USA.
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Michael J Krashes
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA.
- National Institute on Drug Abuse (NIDA), National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
89
|
Inada K, Hagihara M, Tsujimoto K, Abe T, Konno A, Hirai H, Kiyonari H, Miyamichi K. Plasticity of neural connections underlying oxytocin-mediated parental behaviors of male mice. Neuron 2022; 110:2009-2023.e5. [PMID: 35443152 DOI: 10.1016/j.neuron.2022.03.033] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 03/04/2022] [Accepted: 03/28/2022] [Indexed: 11/15/2022]
Abstract
The adult brain can flexibly adapt behaviors to specific life-stage demands. For example, while sexually naive male mice are aggressive to the conspecific young, they start to provide caregiving to infants around the time when their own young are expected. How such behavioral plasticity is implemented at the level of neural connections remains poorly understood. Here, using viral-genetic approaches, we establish hypothalamic oxytocin neurons as the key regulators of the parental caregiving behaviors of male mice. We then use rabies-virus-mediated unbiased screening to identify excitatory neural connections originating from the lateral hypothalamus to the oxytocin neurons to be drastically strengthened when male mice become fathers. These connections are functionally relevant, as their activation suppresses pup-directed aggression in virgin males. These results demonstrate the life-stage associated, long-distance, and cell-type-specific plasticity of neural connections in the hypothalamus, the brain region that is classically assumed to be hard-wired.
Collapse
Affiliation(s)
- Kengo Inada
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan.
| | - Mitsue Hagihara
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
| | - Kazuko Tsujimoto
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
| | - Takaya Abe
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
| | - Ayumu Konno
- Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan; Viral Vector Core, Gunma University Initiative for Advanced Research (GIAR), Maebashi, Gunma 371-8511, Japan
| | - Hirokazu Hirai
- Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan; Viral Vector Core, Gunma University Initiative for Advanced Research (GIAR), Maebashi, Gunma 371-8511, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
| | - Kazunari Miyamichi
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan; Japan Science and Technology Agency, PRESTO, Kawaguchi, Saitama 332-0012, Japan.
| |
Collapse
|
90
|
Watts AG, Kanoski SE, Sanchez-Watts G, Langhans W. The physiological control of eating: signals, neurons, and networks. Physiol Rev 2022; 102:689-813. [PMID: 34486393 PMCID: PMC8759974 DOI: 10.1152/physrev.00028.2020] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
During the past 30 yr, investigating the physiology of eating behaviors has generated a truly vast literature. This is fueled in part by a dramatic increase in obesity and its comorbidities that has coincided with an ever increasing sophistication of genetically based manipulations. These techniques have produced results with a remarkable degree of cell specificity, particularly at the cell signaling level, and have played a lead role in advancing the field. However, putting these findings into a brain-wide context that connects physiological signals and neurons to behavior and somatic physiology requires a thorough consideration of neuronal connections: a field that has also seen an extraordinary technological revolution. Our goal is to present a comprehensive and balanced assessment of how physiological signals associated with energy homeostasis interact at many brain levels to control eating behaviors. A major theme is that these signals engage sets of interacting neural networks throughout the brain that are defined by specific neural connections. We begin by discussing some fundamental concepts, including ones that still engender vigorous debate, that provide the necessary frameworks for understanding how the brain controls meal initiation and termination. These include key word definitions, ATP availability as the pivotal regulated variable in energy homeostasis, neuropeptide signaling, homeostatic and hedonic eating, and meal structure. Within this context, we discuss network models of how key regions in the endbrain (or telencephalon), hypothalamus, hindbrain, medulla, vagus nerve, and spinal cord work together with the gastrointestinal tract to enable the complex motor events that permit animals to eat in diverse situations.
Collapse
Affiliation(s)
- Alan G Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Scott E Kanoski
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Graciela Sanchez-Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Eidgenössische Technische Hochschule-Zürich, Schwerzenbach, Switzerland
| |
Collapse
|
91
|
Anneser L, Gemmer A, Eilers T, Alcantara IC, Loos AY, Ryu S, Schuman EM. The neuropeptide Pth2 modulates social behavior and anxiety in zebrafish. iScience 2022; 25:103868. [PMID: 35243231 PMCID: PMC8861652 DOI: 10.1016/j.isci.2022.103868] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/16/2021] [Accepted: 02/01/2022] [Indexed: 01/09/2023] Open
Abstract
Behavior is context-dependent and often modulated by an animal's internal state. In particular, different social contexts can alter anxiety levels and modulate social behavior. The vertebrate-specific neuropeptide parathyroid hormone 2 (pth2) is regulated by the presence of conspecifics in zebrafish. As its cognate receptor, the parathyroid hormone 2 receptor (pth2r), is widely expressed across the brain, we tested fish lacking the functional Pth2 peptide in several anxiety-related and social behavior paradigms. Here, we show that the propensity to react to sudden stimuli with an escape response was increased in pth2 -/- zebrafish, consistent with an elevated anxiety level. While overall social preference for conspecifics was maintained in pth2 -/- fish until the early juvenile stage, we found that both social preference and shoaling were altered later in development. The data presented suggest that the neuropeptide Pth2 modulates several conserved behaviors and may thus enable the animal to react appropriately in different social contexts.
Collapse
Affiliation(s)
- Lukas Anneser
- Max Planck Institute for Brain Research, Frankfurt am Main 60438, Germany
| | - Anja Gemmer
- Max Planck Institute for Brain Research, Frankfurt am Main 60438, Germany
| | - Tim Eilers
- Max Planck Institute for Brain Research, Frankfurt am Main 60438, Germany
| | - Ivan C. Alcantara
- Max Planck Institute for Brain Research, Frankfurt am Main 60438, Germany
| | - Anett-Yvonn Loos
- Max Planck Institute for Brain Research, Frankfurt am Main 60438, Germany
| | - Soojin Ryu
- Living Systems Institute & College of Medicine and Health, University of Exeter, Exeter EX4 4QD, UK
- Johannes Gutenberg University Medical Center, Mainz 55131, Germany
| | - Erin M. Schuman
- Max Planck Institute for Brain Research, Frankfurt am Main 60438, Germany
| |
Collapse
|
92
|
Neural circuit control of innate behaviors. SCIENCE CHINA. LIFE SCIENCES 2022; 65:466-499. [PMID: 34985643 DOI: 10.1007/s11427-021-2043-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022]
Abstract
All animals possess a plethora of innate behaviors that do not require extensive learning and are fundamental for their survival and propagation. With the advent of newly-developed techniques such as viral tracing and optogenetic and chemogenetic tools, recent studies are gradually unraveling neural circuits underlying different innate behaviors. Here, we summarize current development in our understanding of the neural circuits controlling predation, feeding, male-typical mating, and urination, highlighting the role of genetically defined neurons and their connections in sensory triggering, sensory to motor/motivation transformation, motor/motivation encoding during these different behaviors. Along the way, we discuss possible mechanisms underlying binge-eating disorder and the pro-social effects of the neuropeptide oxytocin, elucidating the clinical relevance of studying neural circuits underlying essential innate functions. Finally, we discuss some exciting brain structures recurrently appearing in the regulation of different behaviors, which suggests both divergence and convergence in the neural encoding of specific innate behaviors. Going forward, we emphasize the importance of multi-angle and cross-species dissections in delineating neural circuits that control innate behaviors.
Collapse
|
93
|
Modulation of itch and pain signals processing in ventrobasal thalamus by thalamic reticular nucleus. iScience 2022; 25:103625. [PMID: 35106466 PMCID: PMC8786640 DOI: 10.1016/j.isci.2021.103625] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/16/2021] [Accepted: 12/10/2021] [Indexed: 01/03/2023] Open
Abstract
Thalamic reticular nucleus (TRN) is known to be crucial for dynamically modulating sensory processing. Recently, the functional role of TRN in itch and pain sensation processing has drawn much attention. We found that ventrobasal thalamus (VB) neurons exhibited scratching behavior-related and nociceptive behavior-related neuronal activity changes, and most of VB neurons responsive to pruritic stimulus were also activated by nociceptive stimulus. Inhibition of VB could relieve itch-induced scratching behaviors and pathological pain without affecting basal nociceptive thresholds, and activation of VB could facilitate scratching behaviors. Tracing and electrophysiology recording results showed that VB mainly received inhibitory inputs from ventral TRN. Furthermore, optogenetic activation of TRN-VB projections suppressed scratching behaviors, and ablation of TRN enhanced scratching behaviors. In addition, activation of TRN-VB projections relieved the pathological pain without affecting basal nociceptive thresholds. Thus, our study indicates that TRN modulates itch and pain signals processing via TRN-VB inhibitory projections. VB is involved in both itch and pain signals processing Manipulation of VB or TRN-VB inhibitory projections modulates both itch and pain Enhancing the inhibitory tone might be a strategy for treating itch and pain
Collapse
|
94
|
Alhadeff AL. A hunger for warmth. Neuron 2022; 110:180-182. [PMID: 35051361 DOI: 10.1016/j.neuron.2021.12.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Humans and animals alike perform behaviors-like putting on a sweater or building a warm nest-to regulate body temperature. In this issue of Neuron, Jung et al. (2022) reveal a parabrachial nucleus-to-lateral hypothalamus circuit that regulates thermoregulatory behavior, a circuit distinct from that which governs motivated feeding behavior.
Collapse
Affiliation(s)
- Amber L Alhadeff
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA; Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
95
|
Kim TH, Schnitzer MJ. Fluorescence imaging of large-scale neural ensemble dynamics. Cell 2022; 185:9-41. [PMID: 34995519 PMCID: PMC8849612 DOI: 10.1016/j.cell.2021.12.007] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 12/14/2022]
Abstract
Recent progress in fluorescence imaging allows neuroscientists to observe the dynamics of thousands of individual neurons, identified genetically or by their connectivity, across multiple brain areas and for extended durations in awake behaving mammals. We discuss advances in fluorescent indicators of neural activity, viral and genetic methods to express these indicators, chronic animal preparations for long-term imaging studies, and microscopes to monitor and manipulate the activity of large neural ensembles. Ca2+ imaging studies of neural activity can track brain area interactions and distributed information processing at cellular resolution. Across smaller spatial scales, high-speed voltage imaging reveals the distinctive spiking patterns and coding properties of targeted neuron types. Collectively, these innovations will propel studies of brain function and dovetail with ongoing neuroscience initiatives to identify new neuron types and develop widely applicable, non-human primate models. The optical toolkit's growing sophistication also suggests that "brain observatory" facilities would be useful open resources for future brain-imaging studies.
Collapse
Affiliation(s)
- Tony Hyun Kim
- James Clark Center for Biomedical Engineering & Sciences, Stanford University, Stanford, CA 94305, USA; CNC Program, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| | - Mark J Schnitzer
- James Clark Center for Biomedical Engineering & Sciences, Stanford University, Stanford, CA 94305, USA; CNC Program, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
96
|
Oxytocin Facilitates Allomaternal Behavior under Stress in Laboratory Mice. eNeuro 2022; 9:ENEURO.0405-21.2022. [PMID: 35017259 PMCID: PMC8868028 DOI: 10.1523/eneuro.0405-21.2022] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/06/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022] Open
Abstract
Oxytocin (Oxt) controls reproductive physiology and various kinds of social behaviors, but the exact contribution of Oxt to different components of parental care still needs to be determined. Here, we illustrate the neuroanatomical relations of the parental nurturing-induced neuronal activation with magnocellular Oxt neurons and fibers in the medial preoptic area (MPOA), the brain region critical for parental and alloparental behaviors. We used genetically-targeted mouse lines for Oxt, Oxt receptor (Oxtr), vasopressin receptor 1a (Avpr1a), vasopressin receptor 1b (Avpr1b), and thyrotropin-releasing hormone (Trh) to systematically examine the role of Oxt-related signaling in pup-directed behaviors. The Oxtr-Avpr1a-Avpr1b triple knock-out (TKO), and Oxt-Trh-Avpr1a-Avpr1b quadruple KO (QKO) mice were grossly healthy and fertile, except for their complete deficiency in milk ejection and modest deficiency in parturition secondary to maternal loss of the Oxt or Oxtr gene. In our minimal stress conditions, pup-directed behaviors in TKO and QKO mothers and fathers, virgin females and males were essentially indistinguishable from those of their littermates with other genotypes. However, Oxtr KO virgin females did show decreased pup retrieval in the pup-exposure assay performed right after restraint stress. This stress vulnerability in the Oxtr KO was abolished by the additional Avpr1b KO. The general stress sensitivity, as measured by plasma cortisol elevation after restraint stress or by the behavioral performance in the open field (OF) and elevated plus maze (EPM), were not altered in the Oxtr KO but were reduced in the Avpr1b KO females, indicating that the balance of neurohypophysial hormones affects the outcome of pup-directed behaviors.
Collapse
|
97
|
Kim DW, Place E, Chinnaiya K, Manning E, Sun C, Dai W, Groves I, Ohyama K, Burbridge S, Placzek M, Blackshaw S. Single-cell analysis of early chick hypothalamic development reveals that hypothalamic cells are induced from prethalamic-like progenitors. Cell Rep 2022; 38:110251. [PMID: 35045288 PMCID: PMC8918062 DOI: 10.1016/j.celrep.2021.110251] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 09/13/2021] [Accepted: 12/20/2021] [Indexed: 01/05/2023] Open
Affiliation(s)
- Dong Won Kim
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elsie Place
- School of Biosciences, University of Sheffield, Sheffield, UK; Bateson Centre, University of Sheffield, Sheffield, UK; Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Kavitha Chinnaiya
- School of Biosciences, University of Sheffield, Sheffield, UK; Bateson Centre, University of Sheffield, Sheffield, UK; Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Elizabeth Manning
- School of Biosciences, University of Sheffield, Sheffield, UK; Bateson Centre, University of Sheffield, Sheffield, UK; Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Changyu Sun
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Weina Dai
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ian Groves
- School of Mathematics and Statistics, University of Sheffield, Sheffield, UK
| | - Kyoji Ohyama
- School of Biosciences, University of Sheffield, Sheffield, UK; Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Sarah Burbridge
- School of Biosciences, University of Sheffield, Sheffield, UK; Bateson Centre, University of Sheffield, Sheffield, UK; Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Marysia Placzek
- School of Biosciences, University of Sheffield, Sheffield, UK; Bateson Centre, University of Sheffield, Sheffield, UK; Neuroscience Institute, University of Sheffield, Sheffield, UK.
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
98
|
Short AK, Thai CW, Chen Y, Kamei N, Pham AL, Birnie MT, Bolton JL, Mortazavi A, Baram TZ. Single-Cell Transcriptional Changes in Hypothalamic Corticotropin-Releasing Factor-Expressing Neurons After Early-Life Adversity Inform Enduring Alterations in Vulnerabilities to Stress. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2021; 3:99-109. [PMID: 36712559 PMCID: PMC9874075 DOI: 10.1016/j.bpsgos.2021.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/11/2021] [Accepted: 12/03/2021] [Indexed: 02/01/2023] Open
Abstract
Background Mental health and vulnerabilities to neuropsychiatric disorders involve the interplay of genes and environment, particularly during sensitive developmental periods. Early-life adversity (ELA) and stress promote vulnerabilities to stress-related affective disorders, yet it is unknown how transient ELA dictates lifelong neuroendocrine and behavioral reactions to stress. The population of hypothalamic corticotropin-releasing factor (CRF)-expressing neurons that regulate stress responses is a promising candidate to mediate the long-lasting influences of ELA on stress-related behavioral and hormonal responses via enduring transcriptional and epigenetic mechanisms. Methods Capitalizing on a well-characterized model of ELA, we examined ELA-induced changes in gene expression profiles of CRF-expressing neurons in the hypothalamic paraventricular nucleus of developing male mice. We used single-cell RNA sequencing on isolated CRF-expressing neurons. We determined the enduring functional consequences of transcriptional changes on stress reactivity in adult ELA mice, including hormonal responses to acute stress, adrenal weights as a measure of chronic stress, and behaviors in the looming shadow threat task. Results Single-cell transcriptomics identified distinct and novel CRF-expressing neuronal populations, characterized by both their gene expression repertoire and their neurotransmitter profiles. ELA-provoked expression changes were selective to specific subpopulations and affected genes involved in neuronal differentiation, synapse formation, energy metabolism, and cellular responses to stress and injury. Importantly, these expression changes were impactful, apparent from adrenal hypertrophy and augmented behavioral responses to stress in adulthood. Conclusions We uncover a novel repertoire of stress-regulating CRF cell types differentially affected by ELA and resulting in augmented stress vulnerability, with relevance to the origins of stress-related affective disorders.
Collapse
Affiliation(s)
- Annabel K. Short
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California,Department of Pediatrics, University of California Irvine, Irvine, California
| | - Christina W. Thai
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California
| | - Yuncai Chen
- Department of Pediatrics, University of California Irvine, Irvine, California
| | - Noriko Kamei
- Department of Pediatrics, University of California Irvine, Irvine, California
| | - Aidan L. Pham
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California,Department of Pediatrics, University of California Irvine, Irvine, California
| | - Matthew T. Birnie
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California,Department of Pediatrics, University of California Irvine, Irvine, California
| | - Jessica L. Bolton
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California,Department of Pediatrics, University of California Irvine, Irvine, California
| | - Ali Mortazavi
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California
| | - Tallie Z. Baram
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California,Department of Pediatrics, University of California Irvine, Irvine, California,Department of Neurology, University of California Irvine, Irvine, California,Address correspondence to Tallie Z. Baram, M.D., Ph.D.
| |
Collapse
|
99
|
Wang Y, Eddison M, Fleishman G, Weigert M, Xu S, Wang T, Rokicki K, Goina C, Henry FE, Lemire AL, Schmidt U, Yang H, Svoboda K, Myers EW, Saalfeld S, Korff W, Sternson SM, Tillberg PW. EASI-FISH for thick tissue defines lateral hypothalamus spatio-molecular organization. Cell 2021; 184:6361-6377.e24. [PMID: 34875226 DOI: 10.1016/j.cell.2021.11.024] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 08/22/2021] [Accepted: 11/12/2021] [Indexed: 11/17/2022]
Abstract
Determining the spatial organization and morphological characteristics of molecularly defined cell types is a major bottleneck for characterizing the architecture underpinning brain function. We developed Expansion-Assisted Iterative Fluorescence In Situ Hybridization (EASI-FISH) to survey gene expression in brain tissue, as well as a turnkey computational pipeline to rapidly process large EASI-FISH image datasets. EASI-FISH was optimized for thick brain sections (300 μm) to facilitate reconstruction of spatio-molecular domains that generalize across brains. Using the EASI-FISH pipeline, we investigated the spatial distribution of dozens of molecularly defined cell types in the lateral hypothalamic area (LHA), a brain region with poorly defined anatomical organization. Mapping cell types in the LHA revealed nine spatially and molecularly defined subregions. EASI-FISH also facilitates iterative reanalysis of scRNA-seq datasets to determine marker-genes that further dissociated spatial and morphological heterogeneity. The EASI-FISH pipeline democratizes mapping molecularly defined cell types, enabling discoveries about brain organization.
Collapse
Affiliation(s)
- Yuhan Wang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Mark Eddison
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Greg Fleishman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Martin Weigert
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; Institute of Bioengineering, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Shengjin Xu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Tim Wang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Konrad Rokicki
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Cristian Goina
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Fredrick E Henry
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Andrew L Lemire
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Uwe Schmidt
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Hui Yang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Karel Svoboda
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Eugene W Myers
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Stephan Saalfeld
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Wyatt Korff
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Scott M Sternson
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA; Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Paul W Tillberg
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA.
| |
Collapse
|
100
|
Leithead AB, Tasker JG, Harony‐Nicolas H. The interplay between glutamatergic circuits and oxytocin neurons in the hypothalamus and its relevance to neurodevelopmental disorders. J Neuroendocrinol 2021; 33:e13061. [PMID: 34786775 PMCID: PMC8951898 DOI: 10.1111/jne.13061] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/14/2021] [Accepted: 10/30/2021] [Indexed: 11/27/2022]
Abstract
Oxytocin (OXT) neurons of the hypothalamus are at the center of several physiological functions, including milk ejection, uterus contraction, and maternal and social behavior. In lactating females, OXT neurons show a pattern of burst firing and inter-neuron synchronization during suckling that leads to pulsatile release of surges of OXT into the bloodstream to stimulate milk ejection. This pattern of firing and population synchronization may be facilitated in part by hypothalamic glutamatergic circuits, as has been observed in vitro using brain slices obtained from male rats and neonates. However, it remains unknown how hypothalamic glutamatergic circuits influence OXT cell activity outside the context of lactation. In this review, we summarize the in vivo and in vitro studies that describe the synchronized burst firing pattern of OXT neurons and the implication of hypothalamic glutamate in this pattern of firing. We also make note of the few studies that have traced glutamatergic afferents to the hypothalamic paraventricular and supraoptic nuclei. Finally, we discuss the genetic findings implicating several glutamatergic genes in neurodevelopmental disorders, including autism spectrum disorder, thus underscoring the need for future studies to investigate the impact of these mutations on hypothalamic glutamatergic circuits and the OXT system.
Collapse
Affiliation(s)
- Amanda B. Leithead
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Seaver Autism Center for Research and TreatmentNew YorkNYUSA
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Friedman Brain Institute at the Icahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Jeffrey G. Tasker
- Neurobiology DivisionDepartment of Cell and Molecular BiologyTulane UniversityNew OrleansLAUSA
| | - Hala Harony‐Nicolas
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Seaver Autism Center for Research and TreatmentNew YorkNYUSA
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Friedman Brain Institute at the Icahn School of Medicine at Mount SinaiNew YorkNYUSA
- Mindich Child Health and Development Institute at the Icahn School of Medicine at Mount SinaiNew YorkNYUSA
| |
Collapse
|