151
|
Allen AT, Heaton EC, Shapiro LP, Butkovich LM, Yount ST, Davies RA, Li DC, Swanson AM, Gourley SL. Inter-individual variability amplified through breeding reveals control of reward-related action strategies by Melanocortin-4 Receptor in the dorsomedial striatum. Commun Biol 2022; 5:116. [PMID: 35136204 PMCID: PMC8825839 DOI: 10.1038/s42003-022-03043-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/06/2022] [Indexed: 12/15/2022] Open
Abstract
In day-to-day life, we often must choose between pursuing familiar behaviors or adjusting behaviors when new strategies might be more fruitful. The dorsomedial striatum (DMS) is indispensable for arbitrating between old and new action strategies. To uncover molecular mechanisms, we trained mice to generate nose poke responses for food, then uncoupled the predictive relationship between one action and its outcome. We then bred the mice that failed to rapidly modify responding. This breeding created offspring with the same tendencies, failing to inhibit behaviors that were not reinforced. These mice had less post-synaptic density protein 95 in the DMS. Also, densities of the melanocortin-4 receptor (MC4R), a high-affinity receptor for α-melanocyte-stimulating hormone, predicted individuals' response strategies. Specifically, high MC4R levels were associated with poor response inhibition. We next found that reducing Mc4r in the DMS in otherwise typical mice expedited response inhibition, allowing mice to modify behavior when rewards were unavailable or lost value. This process required inputs from the orbitofrontal cortex, a brain region canonically associated with response strategy switching. Thus, MC4R in the DMS appears to propel reward-seeking behavior, even when it is not fruitful, while moderating MC4R presence increases the capacity of mice to inhibit such behaviors.
Collapse
Affiliation(s)
- Aylet T Allen
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Elizabeth C Heaton
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Graduate Program in Neuroscience, Emory University, Atlanta, GA, USA
| | - Lauren P Shapiro
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Graduate Program in Molecular and Systems Pharmacology, Emory University, Atlanta, GA, USA
| | - Laura M Butkovich
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Sophie T Yount
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Graduate Program in Molecular and Systems Pharmacology, Emory University, Atlanta, GA, USA
| | - Rachel A Davies
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Dan C Li
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Graduate Program in Neuroscience, Emory University, Atlanta, GA, USA
| | - Andrew M Swanson
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Graduate Program in Neuroscience, Emory University, Atlanta, GA, USA
| | - Shannon L Gourley
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, USA.
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.
- Graduate Program in Neuroscience, Emory University, Atlanta, GA, USA.
- Graduate Program in Molecular and Systems Pharmacology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
152
|
Millard SJ, Bearden CE, Karlsgodt KH, Sharpe MJ. The prediction-error hypothesis of schizophrenia: new data point to circuit-specific changes in dopamine activity. Neuropsychopharmacology 2022; 47:628-640. [PMID: 34588607 PMCID: PMC8782867 DOI: 10.1038/s41386-021-01188-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/23/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023]
Abstract
Schizophrenia is a severe psychiatric disorder affecting 21 million people worldwide. People with schizophrenia suffer from symptoms including psychosis and delusions, apathy, anhedonia, and cognitive deficits. Strikingly, schizophrenia is characterised by a learning paradox involving difficulties learning from rewarding events, whilst simultaneously 'overlearning' about irrelevant or neutral information. While dysfunction in dopaminergic signalling has long been linked to the pathophysiology of schizophrenia, a cohesive framework that accounts for this learning paradox remains elusive. Recently, there has been an explosion of new research investigating how dopamine contributes to reinforcement learning, which illustrates that midbrain dopamine contributes in complex ways to reinforcement learning, not previously envisioned. This new data brings new possibilities for how dopamine signalling contributes to the symptomatology of schizophrenia. Building on recent work, we present a new neural framework for how we might envision specific dopamine circuits contributing to this learning paradox in schizophrenia in the context of models of reinforcement learning. Further, we discuss avenues of preclinical research with the use of cutting-edge neuroscience techniques where aspects of this model may be tested. Ultimately, it is hoped that this review will spur to action more research utilising specific reinforcement learning paradigms in preclinical models of schizophrenia, to reconcile seemingly disparate symptomatology and develop more efficient therapeutics.
Collapse
Affiliation(s)
- Samuel J Millard
- Department of Psychology, University of California, Los Angeles, CA, 90095, USA.
| | - Carrie E Bearden
- Department of Psychology, University of California, Los Angeles, CA, 90095, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, 90095, USA
| | - Katherine H Karlsgodt
- Department of Psychology, University of California, Los Angeles, CA, 90095, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, 90095, USA
| | - Melissa J Sharpe
- Department of Psychology, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
153
|
Shan Q, Hu Y, Chen S, Tian Y. Nucleus accumbens dichotomically controls social dominance in male mice. Neuropsychopharmacology 2022; 47:776-787. [PMID: 34750567 PMCID: PMC8783020 DOI: 10.1038/s41386-021-01220-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 02/05/2023]
Abstract
Social dominance versus social submissiveness is a basic behavioral trait of social animals such as human beings and laboratory mice. The brain regions associated with this behavior have been intensely investigated, and early neuroimaging research on human subjects implies that the nucleus accumbens (NAc) might be involved in encoding social dominance. However, the underlying circuitry and synaptic mechanism are largely unknown. In this study, by introducing lesions to both NAc subregions, the shell and core, a causal relationship is established between social dominance and both NAc subregions. A further electrophysiology investigation on the circuitry of these two subregions revealed that the postsynaptic strength of excitatory synapses onto the medium spiny neurons that express the D1 dopamine receptors in the shell is negatively correlated, and the postsynaptic strength of excitatory synapses onto the medium spiny neurons that express the D2 dopamine receptors in the core is positively correlated, with social dominance. Correspondingly, a DREADD investigation revealed that the activities of these respective medium spiny neurons suppress and promote social dominance. These findings identify a neural substrate for social dominance, implying the potential for a therapeutic strategy for treating related psychiatric disorders.
Collapse
Affiliation(s)
- Qiang Shan
- Laboratory for Synaptic Plasticity, Shantou University Medical College, Shantou, 515041, Guangdong, China.
| | - You Hu
- Laboratory for Synaptic Plasticity, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Shijie Chen
- Laboratory for Synaptic Plasticity, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Yao Tian
- Chern Institute of Mathematics, Nankai University, 300071, Tianjin, China
| |
Collapse
|
154
|
Leighton AH, Victoria Fernández Busch M, Coppens JE, Heimel JA, Lohmann C. Lightweight, wireless LED implant for chronic manipulation in vivo of spontaneous activity in neonatal mice. J Neurosci Methods 2022; 373:109548. [DOI: 10.1016/j.jneumeth.2022.109548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/23/2022] [Accepted: 02/26/2022] [Indexed: 11/27/2022]
|
155
|
Masini D, Kiehn O. Targeted activation of midbrain neurons restores locomotor function in mouse models of parkinsonism. Nat Commun 2022; 13:504. [PMID: 35082287 PMCID: PMC8791953 DOI: 10.1038/s41467-022-28075-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 01/07/2022] [Indexed: 12/26/2022] Open
Abstract
The pedunculopontine nucleus (PPN) is a locomotor command area containing glutamatergic neurons that control locomotor initiation and maintenance. These motor actions are deficient in Parkinson’s disease (PD), where dopaminergic neurodegeneration alters basal ganglia activity. Being downstream of the basal ganglia, the PPN may be a suitable target for ameliorating parkinsonian motor symptoms. Here, we use in vivo cell-type specific PPN activation to restore motor function in two mouse models of parkinsonism made by acute pharmacological blockage of dopamine transmission. With a combination of chemo- and opto-genetics, we show that excitation of caudal glutamatergic PPN neurons can normalize the otherwise severe locomotor deficit in PD, whereas targeting the local GABAergic population only leads to recovery of slow locomotion. The motor rescue driven by glutamatergic PPN activation is independent of activity in nearby locomotor promoting glutamatergic Cuneiform neurons. Our observations point to caudal glutamatergic PPN neurons as a potential target for neuromodulatory restoration of locomotor function in PD. Here, the authors use cell-type specific stimulation of brainstem neurons within the caudal pedunculopontine nucleus to show that activation of excitatory neurons can normalize severe locomotor deficit in mouse models of parkinsonism. The study defines a potential target for neuromodulatory restoration of locomotor function in Parkinson’s disease.
Collapse
Affiliation(s)
- Débora Masini
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Ole Kiehn
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark. .,Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
156
|
Chen Z, Lin MT, Zhan C, Zhong NS, Mu D, Lai KF, Liu MJ. A descending pathway emanating from the periaqueductal gray mediates the development of cough-like hypersensitivity. iScience 2022; 25:103641. [PMID: 35028531 PMCID: PMC8741493 DOI: 10.1016/j.isci.2021.103641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/11/2021] [Accepted: 12/13/2021] [Indexed: 01/10/2023] Open
Abstract
Chronic cough is a common refractory symptom of various respiratory diseases. However, the neural mechanisms that modulate the cough sensitivity and mediate chronic cough remain elusive. Here, we report that GABAergic neurons in the lateral/ventrolateral periaqueductal gray (l/vlPAG) suppress cough processing via a descending pathway. We found that l/vlPAG neurons are activated by coughing-like behaviors and that tussive agent-evoked coughing-like behaviors are impaired after activation of l/vlPAG neurons. In addition, we showed that l/vlPAG neurons form inhibitory synapses with the nucleus of the solitary tract (NTS) neurons. The synaptic strength of these inhibitory projections is weaker in cough hypersensitivity model mice than in naïve mice. Important, activation of l/vlPAG GABAergic neurons projecting to the NTS decreases coughing-like behaviors. In contrast, suppressing these neurons enhances cough sensitivity. These results support the notion that l/vlPAG GABAergic neurons play important roles in cough hypersensitivity and chronic cough through disinhibition of cough processing at the medullary level. GABAergic neurons in the l/vlPAG inhibit coughing-like behaviors The l/vlPAG sends predominately inhibitory projections to the NTS l/vlPAG GABAergic neurons modulate coughing-like behaviors via descending projections l/vlPAG-NTS projections mediate cough hypersensitivity via disinhibitory mechanisms
Collapse
Affiliation(s)
- Zhe Chen
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Xi Road, Guangzhou 510120, China.,Laboratory of Cough, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu 215300, China
| | - Ming-Tong Lin
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Xi Road, Guangzhou 510120, China
| | - Chen Zhan
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Xi Road, Guangzhou 510120, China
| | - Nan-Shan Zhong
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Xi Road, Guangzhou 510120, China
| | - Di Mu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 650 Xin Song Jiang Road, Shanghai 201620, China
| | - Ke-Fang Lai
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Xi Road, Guangzhou 510120, China
| | - Mingzhe J Liu
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Xi Road, Guangzhou 510120, China
| |
Collapse
|
157
|
Senoo A, Yamada Y, Ojima K, Doura T, Hamachi I, Kiyonaka S. Orthogonal Activation of Metabotropic Glutamate Receptor Using Coordination Chemogenetics. Front Chem 2022; 9:825669. [PMID: 35096780 PMCID: PMC8795677 DOI: 10.3389/fchem.2021.825669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/28/2021] [Indexed: 12/01/2022] Open
Abstract
Cell-surface receptors play a pivotal role as transducers of extracellular input. Although different cell types express the same receptor, the physiological roles of the receptor are highly dependent on cell type. To understand each role, tactics for cell-specific activation of the target receptor are in high demand. Herein, we developed an orthogonal activation method targeting metabotropic glutamate receptor 1 (mGlu1), a G-protein coupled receptor. In this method, direct activation via coordination-based chemogenetics (dA-CBC) was adopted, where activation of mGlu1 was artificially induced by a protein conformational change in response to the coordination of a metal ion or metal-ion complex. Our structure-based protein design and screening approach identified mGlu1 mutants that were directly activated by the coordination of Cu2+ or Zn2+, in addition to our previous Pd-complex-sensitive mGlu1 mutant. Notably, the activation of the mutants was mutually orthogonal, resulting in cell-type selective activation in a model system using HEK293 cells.
Collapse
Affiliation(s)
- Akinobu Senoo
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Yutaro Yamada
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Kento Ojima
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Tomohiro Doura
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
- ERATO (Exploratory Research for Advanced Technology, JST), Tokyo, Japan
| | - Shigeki Kiyonaka
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| |
Collapse
|
158
|
Thorens B. Neuronal regulation of glucagon secretion and gluconeogenesis. J Diabetes Investig 2022; 13:599-607. [PMID: 34989155 PMCID: PMC9017634 DOI: 10.1111/jdi.13745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/02/2022] [Indexed: 11/29/2022] Open
Abstract
Hypoglycemia almost never develops in healthy individuals because multiple hypoglycemia sensing systems, located in the periphery and in the central nervous system trigger a coordinated counterregulatory hormonal response to restore normoglycemia. This involves not only the secretion of glucagon but also of epinephrine, norepinephrine, cortisol and growth hormone. Increased hepatic glucose production is also stimulated by direct autonomous nervous connections to the liver that stimulate glycogenolysis and gluconeogenesis. This counterregulatory response, however, becomes deregulated in a significant fraction of diabetic patients that receive insulin therapy. This leads to risk of developing hypoglycemic episodes, of increasing severity, which negatively impact the quality of life of the patients. How hypoglycemia is detected by the central nervous system is being actively investigated. Recent studies using novel molecular biological, optogenetic and chemogenetic techniques, allow the characterization of glucose sensing neurons, the mechanisms of hypoglycemia detection, the neuronal circuits in which they are integrated and the physiological responses they control. This review will discuss recent studies aimed at identifying central hypoglycemia sensing neuronal circuits, how neurons are activated by hypoglycemia, and how they restore normoglycemia.
Collapse
Affiliation(s)
- Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, 1015, Lausanne, Switzerland
| |
Collapse
|
159
|
Steinhorn B, Eroglu E, Michel T. Chemogenetic Approaches to Probe Redox Pathways: Implications for Cardiovascular Pharmacology and Toxicology. Annu Rev Pharmacol Toxicol 2022; 62:551-571. [PMID: 34530645 PMCID: PMC10507364 DOI: 10.1146/annurev-pharmtox-012221-082339] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Chemogenetics refers to experimental systems that dynamically regulate the activity of a recombinant protein by providing or withholding the protein's specific biochemical stimulus. Chemogenetic tools permit precise dynamic control of specific signaling molecules to delineate the roles of those molecules in physiology and disease. Yeast d-amino acid oxidase (DAAO) enables chemogenetic manipulation of intracellular redox balance by generating hydrogen peroxide only in the presence of d-amino acids. Advances in biosensors have allowed the precise quantitation of these signaling molecules. The combination of chemogenetic approaches with biosensor methodologies has opened up new lines of investigation, allowing the analysis of intracellular redox pathways that modulate physiological and pathological cell responses. We anticipate that newly developed transgenic chemogenetic models will permit dynamic modulation of cellularredox balance in diverse cells and tissues and will facilitate the identification and validation of novel therapeutic targets involved in both physiological redox pathways and pathological oxidative stress.
Collapse
Affiliation(s)
- Benjamin Steinhorn
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Emrah Eroglu
- Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
- Department of Molecular Biology and Biochemistry, Medical University of Graz, 8036 Graz, Austria
| | - Thomas Michel
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA;
| |
Collapse
|
160
|
Desloovere J, Boon P, Larsen LE, Goossens MG, Delbeke J, Carrette E, Wadman W, Vonck K, Raedt R. Chemogenetic Seizure Control with Clozapine and the Novel Ligand JHU37160 Outperforms the Effects of Levetiracetam in the Intrahippocampal Kainic Acid Mouse Model. Neurotherapeutics 2022; 19:342-351. [PMID: 34862591 PMCID: PMC9130374 DOI: 10.1007/s13311-021-01160-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2021] [Indexed: 01/03/2023] Open
Abstract
Expression of inhibitory designer receptors exclusively activated by designer drugs (DREADDs) on excitatory hippocampal neurons in the hippocampus represents a potential new therapeutic strategy for drug-resistant epilepsy. To overcome the limitations of the commonly used DREADD agonist clozapine, we investigated the efficacy of the novel DREADD ligand JHU37160 in chemogenetic seizure suppression in the intrahippocampal kainic acid (IHKA) mouse model for temporal lobe epilepsy (TLE). In addition, seizure-suppressing effects of chemogenetics were compared to the commonly used anti-epileptic drug (AED), levetiracetam (LEV). Therefore, an adeno-associated viral vector was injected in the sclerotic hippocampus of IHKA mice to induce expression of a tagged inhibitory DREADD hM4Di or only a tag (control) specifically in excitatory neurons using the CamKIIα promoter. Subsequently, animals were treated with LEV (800 mg/kg), clozapine (0.1 mg/kg), and DREADD ligand JHU37160 (0.1 mg/kg) and the effect on spontaneous seizures was investigated. Clozapine and JHU37160-mediated chemogenetic treatment both suppressed seizures in DREADD-expressing IHKA mice. Clozapine treatment suppressed seizures up to 34 h after treatment, and JHU37160 effects lasted for 26 h after injection. Moreover, both compounds reduced the length of seizures that did occur after treatment up to 28 h and 18 h after clozapine and JHU37160, respectively. No seizure-suppressing effects were found in control animals using these ligands. Chemogenetic seizure treatment suppressed seizures during the first 30 min after injection, and seizures remained suppressed during 8 h following treatment. Chemogenetics thus outperformed effects of levetiracetam (p < 0.001), which suppressed seizure frequency with a maximum of 55 ± 9% for up to 1.5 h (p < 0.05). Only chemogenetic and not levetiracetam treatment affected the length of seizures after treatment (p < 0.001). These results show that the chemogenetic therapeutic strategy with either clozapine or JHU37160 effectively suppresses spontaneous seizures in the IHKA mouse model, confirming JHU37160 as an effective DREADD ligand. Moreover, chemogenetic therapy outperforms the effects of levetiracetam, indicating its potential to suppress drug-resistant seizures.
Collapse
Affiliation(s)
- Jana Desloovere
- 4BRAIN, Department of Neurology, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Paul Boon
- 4BRAIN, Department of Neurology, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Lars Emil Larsen
- 4BRAIN, Department of Neurology, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
- Medical Image and Signal Processing, Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | | | - Jean Delbeke
- 4BRAIN, Department of Neurology, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Evelien Carrette
- 4BRAIN, Department of Neurology, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Wytse Wadman
- 4BRAIN, Department of Neurology, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Kristl Vonck
- 4BRAIN, Department of Neurology, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Robrecht Raedt
- 4BRAIN, Department of Neurology, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium.
| |
Collapse
|
161
|
Gourley SL, Srikanth KD, Woon EP, Gil-Henn H. Pyk2 Stabilizes Striatal Medium Spiny Neuron Structure and Striatal-Dependent Action. Cells 2021; 10:3442. [PMID: 34943950 PMCID: PMC8700592 DOI: 10.3390/cells10123442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 11/17/2021] [Accepted: 11/30/2021] [Indexed: 01/18/2023] Open
Abstract
In day-to-day life, we often choose between pursuing familiar behaviors that have been rewarded in the past or adjusting behaviors when new strategies might be more fruitful. The dorsomedial striatum (DMS) is indispensable for flexibly arbitrating between old and new behavioral strategies. The way in which DMS neurons host stable connections necessary for sustained flexibility is still being defined. An entry point to addressing this question may be the structural scaffolds on DMS neurons that house synaptic connections. We find that the non-receptor tyrosine kinase Proline-rich tyrosine kinase 2 (Pyk2) stabilizes both dendrites and spines on striatal medium spiny neurons, such that Pyk2 loss causes dendrite arbor and spine loss. Viral-mediated Pyk2 silencing in the DMS obstructs the ability of mice to arbitrate between rewarded and non-rewarded behaviors. Meanwhile, the overexpression of Pyk2 or the closely related focal adhesion kinase (FAK) enhances this ability. Finally, experiments using combinatorial viral vector strategies suggest that flexible, Pyk2-dependent action involves inputs from the medial prefrontal cortex (mPFC), but not the ventrolateral orbitofrontal cortex (OFC). Thus, Pyk2 stabilizes the striatal medium spiny neuron structure, likely providing substrates for inputs, and supports the capacity of mice to arbitrate between novel and familiar behaviors, including via interactions with the medial-prefrontal cortex.
Collapse
Affiliation(s)
- Shannon L. Gourley
- Yerkes National Primate Research Center, Department of Pediatrics, Emory University School of Medicine, 954 Gatewood Rd. NE, Atlanta, GA 30329, USA;
| | - Kolluru D. Srikanth
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel;
| | - Ellen P. Woon
- Yerkes National Primate Research Center, Department of Pediatrics, Emory University School of Medicine, 954 Gatewood Rd. NE, Atlanta, GA 30329, USA;
| | - Hava Gil-Henn
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel;
| |
Collapse
|
162
|
Malfait F, Colman M, Vroman R, De Wandele I, Rombaut L, Miller RE, Malfait AM, Syx D. Pain in the Ehlers-Danlos syndromes: Mechanisms, models, and challenges. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2021; 187:429-445. [PMID: 34797601 DOI: 10.1002/ajmg.c.31950] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/17/2021] [Accepted: 10/22/2021] [Indexed: 12/27/2022]
Abstract
Chronic pain is one of the most common, yet poorly studied, complaints in people suffering from Ehlers-Danlos syndromes (EDS). This heterogeneous group of heritable connective tissue disorders is typically characterized by skin hyperextensibility, joint hypermobility, and generalized connective tissue fragility. Most EDS types are caused by genetic defects that affect connective tissue biosynthesis, thereby compromising collagen biosynthesis or fibrillogenesis and resulting in a disorganized extracellular matrix. Even though chronic pain is a major source of disability, functional impairment, and psychosocial suffering in EDS, currently used analgesics and other treatment strategies provide inadequate pain relief and thus represents an important unmet medical need. An important contributor to this is the lack of knowledge about the underlying mechanisms. In this narrative review, we summarize the current understanding of pain and the associated mechanisms in EDS based on clinical studies focusing on questionnaires and experimental pain testing as well as studies in animal models of EDS. In addition, we highlight the challenges, gaps, and opportunities in EDS-pain research.
Collapse
Affiliation(s)
- Fransiska Malfait
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Marlies Colman
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Robin Vroman
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Inge De Wandele
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Lies Rombaut
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Rachel E Miller
- Division of Rheumatology, Rush University Medical Center, Chicago, Illinois, USA
| | - Anne-Marie Malfait
- Division of Rheumatology, Rush University Medical Center, Chicago, Illinois, USA
| | - Delfien Syx
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
163
|
Kim J, Rahman MH, Lee WH, Suk K. Chemogenetic stimulation of the G i pathway in astrocytes suppresses neuroinflammation. Pharmacol Res Perspect 2021; 9:e00822. [PMID: 34676988 PMCID: PMC8532135 DOI: 10.1002/prp2.822] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/30/2021] [Indexed: 11/06/2022] Open
Abstract
Engineered G protein-coupled receptors (GPCRs) are commonly used in chemogenetics as designer receptors exclusively activated by designer drugs (DREADDs). Although several GPCRs have been studied in astrocytes using a chemogenetic approach, the functional role of the astrocytic Gi pathway is not clear, as the literature is conflicting depending on the brain regions or behaviors investigated. In this study, we evaluated the role of the astrocytic Gi pathway in neuroinflammation using a Gi -coupled DREADD (hM4Di). Gi -DREADD was expressed in hippocampal astrocytes of a lipopolysaccharide (LPS)-induced neuroinflammation mouse model using adeno-associated viruses. We found that astrocyte Gi -DREADD stimulation using clozapine N-oxide (CNO) inhibits neuroinflammation, as characterized by decreased levels of proinflammatory cytokines, glial activation, and cognitive impairment in mice. Subsequent experiments using primary astrocyte cultures revealed that Gi -DREADD stimulation significantly downregulated LPS-induced expression of Nos2 mRNA and nitric oxide production. Similarly, in vitro calcium imaging showed that activation of the astrocytic Gi pathway attenuated intracellular calcium transients triggered by LPS treatment, suggesting a positive correlation between enhanced calcium transients and the inflammatory phenotype of astrocytes observed in the inflamed brain. Taken together, our results indicate that the astrocytic Gi pathway plays an inhibitory role in neuroinflammation, providing an opportunity to identify potential cellular and molecular targets to control neuroinflammation.
Collapse
Affiliation(s)
- Jae‐Hong Kim
- Department of PharmacologySchool of MedicineKyungpook National UniversityDaeguRepublic of Korea
- BK21 Plus KNU Biomedical Convergence ProgramDepartment of Biomedical SciencesSchool of MedicineKyungpook National UniversityDaeguRepublic of Korea
| | - Md Habibur Rahman
- Department of PharmacologySchool of MedicineKyungpook National UniversityDaeguRepublic of Korea
- BK21 Plus KNU Biomedical Convergence ProgramDepartment of Biomedical SciencesSchool of MedicineKyungpook National UniversityDaeguRepublic of Korea
- Brain Science & Engineering InstituteKyungpook National UniversityDaeguRepublic of Korea
| | - Won Ha Lee
- School of Life SciencesBrain Korea 21 Plus KNU Creative BioResearch GroupKyungpook National UniversityDaeguRepublic of Korea
| | - Kyoungho Suk
- Department of PharmacologySchool of MedicineKyungpook National UniversityDaeguRepublic of Korea
- BK21 Plus KNU Biomedical Convergence ProgramDepartment of Biomedical SciencesSchool of MedicineKyungpook National UniversityDaeguRepublic of Korea
- Brain Science & Engineering InstituteKyungpook National UniversityDaeguRepublic of Korea
| |
Collapse
|
164
|
Javdan SB, Deans TL. Design and development of engineered receptors for cell and tissue engineering. CURRENT OPINION IN SYSTEMS BIOLOGY 2021; 28:100363. [PMID: 34527831 PMCID: PMC8437148 DOI: 10.1016/j.coisb.2021.100363] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Advances in synthetic biology have provided genetic tools to reprogram cells to obtain desired cellular functions that include tools to enable the customization of cells to sense an extracellular signal and respond with a desired output. These include a variety of engineered receptors capable of transmembrane signaling that transmit information from outside of the cell to inside when specific ligands bind to them. Recent advances in synthetic receptor engineering have enabled the reprogramming of cell and tissue behavior, controlling cell fate decisions, and providing new vehicles for therapeutic delivery.
Collapse
Affiliation(s)
- Shwan B. Javdan
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT
| | - Tara L. Deans
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT
| |
Collapse
|
165
|
Waldeck-Weiermair M, Yadav S, Spyropoulos F, Krüger C, Pandey AK, Michel T. Dissecting in vivo and in vitro redox responses using chemogenetics. Free Radic Biol Med 2021; 177:360-369. [PMID: 34752919 PMCID: PMC8639655 DOI: 10.1016/j.freeradbiomed.2021.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/08/2021] [Accepted: 11/04/2021] [Indexed: 02/03/2023]
Abstract
Hydrogen peroxide (H2O2) is the most abundant reactive oxygen species (ROS) within mammalian cells. At low concentrations, H2O2 serves as a versatile cell signaling molecule that mediates vital physiological functions. Yet at higher concentrations, H2O2 can be a toxic molecule by promoting pathological oxidative stress in cells and tissues. Within normal cells, H2O2 is differentially distributed in a variety of subcellular locales. Moreover, many redox-active enzymes and their substrates are themselves differentially distributed within cells. Numerous reports have described the biological and biochemical consequences of adding exogenous H2O2 to cultured cells and tissues, but many of these observations are difficult to interpret: the effects of exogenous H2O2 do not necessarily replicate the cellular responses to endogenous H2O2. In recent years, chemogenetic approaches have been developed to dynamically regulate the abundance of H2O2 in specific subcellular locales. Chemogenetic approaches have been applied in multiple experimental systems, ranging from in vitro studies on the intracellular transport and metabolism of H2O2, all the way to in vivo studies that generate oxidative stress in specific organs in living animals. These chemogenetic approaches have exploited a yeast-derived d-amino acid oxidase (DAAO) that synthesizes H2O2 only in the presence of its d-amino acid substrate. DAAO can be targeted to various subcellular locales, and can be dynamically activated by the addition or withdrawal of its d-amino acid substrate. In addition, recent advances in the development of highly sensitive genetically encoded H2O2 biosensors are providing a better understanding of both physiological and pathological oxidative pathways. This review highlights several applications of DAAO as a chemogenetic tool across a wide range of biological systems, from analyses of subcellular H2O2 metabolism in cells to the development of new disease models caused by oxidative stress in vivo.
Collapse
Affiliation(s)
- Markus Waldeck-Weiermair
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA; Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010, Graz, Austria
| | - Shambhu Yadav
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Fotios Spyropoulos
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA; Department of Pediatric Newborn Medicine, Harvard Medical School, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, USA
| | - Christina Krüger
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Arvind K Pandey
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Thomas Michel
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.
| |
Collapse
|
166
|
Xu C, Gong Y, Wang Y, Chen Z. New advances in pharmacoresistant epilepsy towards precise management-from prognosis to treatments. Pharmacol Ther 2021; 233:108026. [PMID: 34718071 DOI: 10.1016/j.pharmthera.2021.108026] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/15/2021] [Accepted: 10/25/2021] [Indexed: 12/13/2022]
Abstract
Epilepsy, one of the most severe neurological diseases, is characterized by abrupt recurrent seizures. Despite great progress in the development of antiseizure drugs (ASDs) based on diverse molecular targets, more than one third of epilepsy patients still show resistance to ASDs, a condition termed pharmacoresistant epilepsy. The management of pharmacoresistant epilepsy involves serious challenges. In the past decade, promising advances have been made in the use of interdisciplinary techniques involving biophysics, bioinformatics, biomaterials and biochemistry, which allow more precise prognosis and development of drug target for pharmacoresistant epilepsy. Notably, novel experimental tools such as viral vector gene delivery, optogenetics and chemogenetics have provided a framework for promising approaches to the precise treatment of pharmacoresistant epilepsy. In this review, historical achievements especially recent advances of the past decade in the prognosis and treatment of pharmacoresistant epilepsy from both clinical and laboratory settings are presented and summarized. We propose that the further development of novel experimental tools at cellular or molecular levels with both temporal and spatial precision are necessary to make improve the management and drug development for pharmacoresistant epilepsy in the clinical arena.
Collapse
Affiliation(s)
- Cenglin Xu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiwei Gong
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China; Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China; Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Epilepsy Center, Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
167
|
Level of hM4D(Gi) DREADD Expression Determines Inhibitory and Neurotoxic Effects in the Hippocampus. eNeuro 2021; 8:ENEURO.0105-21.2021. [PMID: 34620623 PMCID: PMC8570686 DOI: 10.1523/eneuro.0105-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
Selective neuromodulation using designer receptors exclusively activated by designer drugs (DREADDs) has become an increasingly important research tool, as well as an emerging therapeutic approach. However, the safety profile of DREADD expression is unknown. Here, different titers of adeno-associated viral (AAV) vector were administered in an attempt to vary total expression levels of the inhibitory DREADD hM4D(Gi) in excitatory hippocampal neurons. Male Sprague Dawley rats were injected with AAV2/7 encoding DREADD-mCherry, DREADD, or mCherry. Pronounced neuronal loss and neuroinflammatory reactions were observed after transduction with the high titer DREADD AAV, which also resulted in the highest DREADD expression levels. No such effects were observed in the mCherry control group, despite an equally high titer, nor in conditions where lower viral vector titers were injected. In the high titer DREADD conditions, dentate gyrus (DG) evoked potentials were inhibited on clozapine-induced activation of hM4D(Gi), while in low titer conditions DG evoked potentials were enhanced. Recordings of single neuronal activity nevertheless indicated a reduction in spontaneous firing of granule cell layer neurons. Our results indicate that prolonged, high levels of DREADD expression can have neurotoxic effects and that chemogenetic suppression of excitatory hippocampal neurons can paradoxically enhance DG evoked potentials.
Collapse
|
168
|
Axonal CB1 Receptors Mediate Inhibitory Bouton Formation via cAMP Increase and PKA. J Neurosci 2021; 41:8279-8296. [PMID: 34413209 DOI: 10.1523/jneurosci.0851-21.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/30/2021] [Accepted: 07/25/2021] [Indexed: 12/11/2022] Open
Abstract
Experience-dependent formation and removal of inhibitory synapses are essential throughout life. For instance, GABAergic synapses are removed to facilitate learning, and strong excitatory activity is accompanied by the formation of inhibitory synapses to maintain coordination between excitation and inhibition. We recently discovered that active dendrites trigger the growth of inhibitory synapses via CB1 receptor-mediated endocannabinoid signaling, but the underlying mechanism remained unclear. Using two-photon microscopy to monitor the formation of individual inhibitory boutons in hippocampal organotypic slices from mice (both sexes), we found that CB1 receptor activation mediated the formation of inhibitory boutons and promoted their subsequent stabilization. Inhibitory bouton formation did not require neuronal activity and was independent of Gi/o-protein signaling, but was directly induced by elevating cAMP levels using forskolin and by activating Gs-proteins using DREADDs. Blocking PKA activity prevented CB1 receptor-mediated inhibitory bouton formation. Our findings reveal that axonal CB1 receptors signal via unconventional downstream pathways and that inhibitory bouton formation is triggered by an increase in axonal cAMP levels. Our results demonstrate an unexpected role for axonal CB1 receptors in axon-specific, and context-dependent, inhibitory synapse formation.SIGNIFICANCE STATEMENT Coordination between excitation and inhibition is required for proper brain function throughout life. It was previously shown that new inhibitory synapses can be formed in response to strong excitation to maintain this coordination, and this was mediated by endocannabinoid signaling via CB1 receptors. As activation of CB1 receptors generally results in the suppression of synaptic transmission, it remained unclear how CB1 receptors can mediate the formation of inhibitory synapses. Here we show that CB1 receptors on inhibitory axons signal via unconventional intracellular pathways and that inhibitory bouton formation is triggered by an increase in axonal cAMP levels and requires PKA activity. Our findings point to a central role for axonal cAMP signaling in activity-dependent inhibitory synapse formation.
Collapse
|
169
|
Lentini C, d'Orange M, Marichal N, Trottmann MM, Vignoles R, Foucault L, Verrier C, Massera C, Raineteau O, Conzelmann KK, Rival-Gervier S, Depaulis A, Berninger B, Heinrich C. Reprogramming reactive glia into interneurons reduces chronic seizure activity in a mouse model of mesial temporal lobe epilepsy. Cell Stem Cell 2021; 28:2104-2121.e10. [PMID: 34592167 PMCID: PMC8657801 DOI: 10.1016/j.stem.2021.09.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 07/20/2021] [Accepted: 09/03/2021] [Indexed: 12/03/2022]
Abstract
Reprogramming brain-resident glial cells into clinically relevant induced neurons (iNs) is an emerging strategy toward replacing lost neurons and restoring lost brain functions. A fundamental question is now whether iNs can promote functional recovery in pathological contexts. We addressed this question in the context of therapy-resistant mesial temporal lobe epilepsy (MTLE), which is associated with hippocampal seizures and degeneration of hippocampal GABAergic interneurons. Using a MTLE mouse model, we show that retrovirus-driven expression of Ascl1 and Dlx2 in reactive hippocampal glia in situ, or in cortical astroglia grafted in the epileptic hippocampus, causes efficient reprogramming into iNs exhibiting hallmarks of interneurons. These induced interneurons functionally integrate into epileptic networks and establish GABAergic synapses onto dentate granule cells. MTLE mice with GABAergic iNs show a significant reduction in both the number and cumulative duration of spontaneous recurrent hippocampal seizures. Thus glia-to-neuron reprogramming is a potential disease-modifying strategy to reduce seizures in therapy-resistant epilepsy. Retroviruses target reactive hippocampal glia proliferating in a mouse model of mesial temporal lobe epilepsy Ascl1 and Dlx2 reprogram reactive glia into GABAergic interneurons in the epileptic hippocampus Induced interneurons establish GABAergic synapses onto dentate granule cells Induced interneurons reduce chronic epileptic activity in the hippocampus
Collapse
Affiliation(s)
- Célia Lentini
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Marie d'Orange
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Nicolás Marichal
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Marie-Madeleine Trottmann
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Rory Vignoles
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Louis Foucault
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Charlotte Verrier
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Céline Massera
- Univ Grenoble Alpes, Inserm U1216, Grenoble Institut des Neurosciences, 38000 Grenoble, France
| | - Olivier Raineteau
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Karl-Klaus Conzelmann
- Max von Pettenkofer-Institute Virology, Medical Faculty & Gene Center, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Sylvie Rival-Gervier
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, INRAE, Stem Cell and Brain Research Institute U1208, CSC USC1361, 69500 Bron, France
| | - Antoine Depaulis
- Univ Grenoble Alpes, Inserm U1216, Grenoble Institut des Neurosciences, 38000 Grenoble, France
| | - Benedikt Berninger
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK; Institute of Physiological Chemistry, University Medical Center, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Christophe Heinrich
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France.
| |
Collapse
|
170
|
Dominguez-Paredes D, Jahanshahi A, Kozielski KL. Translational considerations for the design of untethered nanomaterials in human neural stimulation. Brain Stimul 2021; 14:1285-1297. [PMID: 34375694 DOI: 10.1016/j.brs.2021.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 07/03/2021] [Accepted: 08/01/2021] [Indexed: 12/18/2022] Open
Abstract
Neural stimulation is a powerful tool to study brain physiology and an effective treatment for many neurological disorders. Conventional interfaces use electrodes implanted in the brain. As these are often invasive and have limited spatial targeting, they carry a potential risk of side-effects. Smaller neural devices may overcome these obstacles, and as such, the field of nanoscale and remotely powered neural stimulation devices is growing. This review will report on current untethered, injectable nanomaterial technologies intended for neural stimulation, with a focus on material-tissue interface engineering. We will review nanomaterials capable of wireless neural stimulation, and discuss their stimulation mechanisms. Taking cues from more established nanomaterial fields (e.g., cancer theranostics, drug delivery), we will then discuss methods to modify material interfaces with passive and bioactive coatings. We will discuss methods of delivery to a desired brain region, particularly in the context of how delivery and localization are affected by surface modification. We will also consider each of these aspects of nanoscale neurostimulators with a focus on their prospects for translation to clinical use.
Collapse
Affiliation(s)
- David Dominguez-Paredes
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Ali Jahanshahi
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Kristen L Kozielski
- Department of Bioengineering and Biosystems, Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany; Department of Electrical and Computer Engineering, Technical University of Munich, Munich, Germany.
| |
Collapse
|
171
|
Donthamsetti P, Winter N, Hoagland A, Stanley C, Visel M, Lammel S, Trauner D, Isacoff E. Cell specific photoswitchable agonist for reversible control of endogenous dopamine receptors. Nat Commun 2021; 12:4775. [PMID: 34362914 PMCID: PMC8346604 DOI: 10.1038/s41467-021-25003-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 07/15/2021] [Indexed: 02/07/2023] Open
Abstract
Dopamine controls diverse behaviors and their dysregulation contributes to many disorders. Our ability to understand and manipulate the function of dopamine is limited by the heterogenous nature of dopaminergic projections, the diversity of neurons that are regulated by dopamine, the varying distribution of the five dopamine receptors (DARs), and the complex dynamics of dopamine release. In order to improve our ability to specifically modulate distinct DARs, here we develop a photo-pharmacological strategy using a Membrane anchored Photoswitchable orthogonal remotely tethered agonist for the Dopamine receptor (MP-D). Our design selectively targets D1R/D5R receptor subtypes, most potently D1R (MP-D1ago), as shown in HEK293T cells. In vivo, we targeted dorsal striatal medium spiny neurons where the photo-activation of MP-D1ago increased movement initiation, although further work is required to assess the effects of MP-D1ago on neuronal function. Our method combines ligand and cell type-specificity with temporally precise and reversible activation of D1R to control specific aspects of movement. Our results provide a template for analyzing dopamine receptors.
Collapse
Affiliation(s)
- Prashant Donthamsetti
- grid.47840.3f0000 0001 2181 7878Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | - Nils Winter
- grid.5252.00000 0004 1936 973XDepartment of Chemistry, Ludwig-Maximilians University, München, Germany
| | - Adam Hoagland
- grid.47840.3f0000 0001 2181 7878Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | - Cherise Stanley
- grid.47840.3f0000 0001 2181 7878Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | - Meike Visel
- grid.47840.3f0000 0001 2181 7878Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | - Stephan Lammel
- grid.47840.3f0000 0001 2181 7878Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | - Dirk Trauner
- grid.137628.90000 0004 1936 8753Department of Chemistry, New York University, New York City, NY USA
| | - Ehud Isacoff
- grid.47840.3f0000 0001 2181 7878Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA ,grid.47840.3f0000 0001 2181 7878Helen Wills Neuroscience Institute, University of California, Berkeley, CA USA ,grid.184769.50000 0001 2231 4551Molecular Biophysics & Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA USA
| |
Collapse
|
172
|
Guo J, Kent A, Davila E. Chimeric non-antigen receptors in T cell-based cancer therapy. J Immunother Cancer 2021; 9:jitc-2021-002628. [PMID: 34344725 PMCID: PMC8336119 DOI: 10.1136/jitc-2021-002628] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2021] [Indexed: 01/04/2023] Open
Abstract
Adoptively transferred T cell-based cancer therapies have shown incredible promise in treatment of various cancers. So far therapeutic strategies using T cells have focused on manipulation of the antigen-recognition machinery itself, such as through selective expression of tumor-antigen specific T cell receptors or engineered antigen-recognition chimeric antigen receptors (CARs). While several CARs have been approved for treatment of hematopoietic malignancies, this kind of therapy has been less successful in the treatment of solid tumors, in part due to lack of suitable tumor-specific targets, the immunosuppressive tumor microenvironment, and the inability of adoptively transferred cells to maintain their therapeutic potentials. It is critical for therapeutic T cells to overcome immunosuppressive environmental triggers, mediating balanced antitumor immunity without causing unwanted inflammation or autoimmunity. To address these hurdles, chimeric receptors with distinct signaling properties are being engineered to function as allies of tumor antigen-specific receptors, modulating unique aspects of T cell function without directly binding to antigen themselves. In this review, we focus on the design and function of these chimeric non-antigen receptors, which fall into three broad categories: ‘inhibitory-to-stimulatory’ switch receptors that bind natural ligands, enhanced stimulatory receptors that interact with natural ligands, and synthetic receptor-ligand pairs. Our intent is to offer detailed descriptions that will help readers to understand the structure and function of these receptors, as well as inspire development of additional novel synthetic receptors to improve T cell-based cancer therapy.
Collapse
Affiliation(s)
- Jitao Guo
- Division of Medical Oncology, Department of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, Colorado, USA
| | - Andrew Kent
- Division of Medical Oncology, Department of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, Colorado, USA
| | - Eduardo Davila
- Division of Medical Oncology, Department of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, Colorado, USA .,Human Immunology and Immunotherapy Initiative, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA.,University of Colorado Comprehensive Cancer Center, Aurora, Colorado, USA.,Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
173
|
Surmeier DJ. Beyond Just Connectivity - Neuronal Activity Drives α-Synuclein Pathology. Mov Disord 2021; 36:1487-1488. [PMID: 34302385 DOI: 10.1002/mds.28618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 11/06/2022] Open
Affiliation(s)
- Dalton James Surmeier
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
174
|
Kim S, Park D, Kim J, Kim D, Kim H, Mori T, Jung H, Lee D, Hong S, Jeon J, Tabuchi K, Cheong E, Kim J, Um JW, Ko J. Npas4 regulates IQSEC3 expression in hippocampal somatostatin interneurons to mediate anxiety-like behavior. Cell Rep 2021; 36:109417. [PMID: 34289353 DOI: 10.1016/j.celrep.2021.109417] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 02/11/2021] [Accepted: 06/28/2021] [Indexed: 02/08/2023] Open
Abstract
Activity-dependent GABAergic synapse plasticity is important for normal brain functions, but the underlying molecular mechanisms remain incompletely understood. Here, we show that Npas4 (neuronal PAS-domain protein 4) transcriptionally regulates the expression of IQSEC3, a GABAergic synapse-specific guanine nucleotide-exchange factor for ADP-ribosylation factor (ARF-GEF) that directly interacts with gephyrin. Neuronal activation by an enriched environment induces Npas4-mediated upregulation of IQSEC3 protein specifically in CA1 stratum oriens layer somatostatin (SST)-expressing GABAergic interneurons. SST+ interneuron-specific knockout (KO) of Npas4 compromises synaptic transmission in these GABAergic interneurons, increases neuronal activity in CA1 pyramidal neurons, and reduces anxiety behavior, all of which are normalized by the expression of wild-type IQSEC3, but not a dominant-negative ARF-GEF-inactive mutant, in SST+ interneurons of Npas4-KO mice. Our results suggest that IQSEC3 is a key GABAergic synapse component that is directed by Npas4 and ARF activity, specifically in SST+ interneurons, to orchestrate excitation-to-inhibition balance and control anxiety-like behavior.
Collapse
Affiliation(s)
- Seungjoon Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Dongseok Park
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Jinhu Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Dongwook Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Hyeonho Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Takuma Mori
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano 390-86221, Japan
| | - Hyeji Jung
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Dongsu Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Sookyung Hong
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Jongcheol Jeon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Katsuhiko Tabuchi
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano 390-86221, Japan; Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano 390-8621, Japan
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Jaehoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Ji Won Um
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea.
| | - Jaewon Ko
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea.
| |
Collapse
|
175
|
Astrocytes in the Ventromedial Hypothalamus Involve Chronic Stress-Induced Anxiety and Bone Loss in Mice. Neural Plast 2021; 2021:7806370. [PMID: 34306063 PMCID: PMC8282369 DOI: 10.1155/2021/7806370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 06/22/2021] [Indexed: 11/18/2022] Open
Abstract
Chronic stress is one of the main risk factors of bone loss. While the neurons and neural circuits of the ventromedial hypothalamus (VMH) mediate bone loss induced by chronic stress, the detailed intrinsic mechanisms within the VMH nucleus still need to be explored. Astrocytes in brain regions play important roles in the regulation of metabolism and anxiety-like behavior through interactions with surrounding neurons. However, whether astrocytes in the VMH affect neuronal activity and therefore regulate chronic stress-induced anxiety and bone loss remain elusive. In this study, we found that VMH astrocytes were activated during chronic stress-induced anxiety and bone loss. Pharmacogenetic activation of the Gi and Gq pathways in VMH astrocytes reduced and increased the levels of anxiety and bone loss, respectively. Furthermore, activation of VMH astrocytes by optogenetics induced depolarization in neighboring steroidogenic factor-1 (SF-1) neurons, which was diminished by administration of N-methyl-D-aspartic acid (NMDA) receptor blocker but not by alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor blocker. These results suggest that there may be a functional "glial-neuron microcircuit" in VMH nuclei that mediates anxiety and bone loss induced by chronic stress. This study not only advances our understanding of glial cell function but also provides a potential intervention target for chronic stress-induced anxiety and bone loss therapy.
Collapse
|
176
|
Koralek AC, Costa RM. Dichotomous dopaminergic and noradrenergic neural states mediate distinct aspects of exploitative behavioral states. SCIENCE ADVANCES 2021; 7:7/30/eabh2059. [PMID: 34301604 PMCID: PMC8302134 DOI: 10.1126/sciadv.abh2059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 06/07/2021] [Indexed: 06/13/2023]
Abstract
The balance between exploiting known actions and exploring alternatives is critical for survival and hypothesized to rely on shifts in neuromodulation. We developed a behavioral paradigm to capture exploitative and exploratory states and imaged calcium dynamics in genetically identified dopaminergic and noradrenergic neurons. During exploitative states, characterized by motivated repetition of the same action choice, dopamine neurons in SNc encoding movement vigor showed sustained elevation of basal activity that lasted many seconds. This sustained activity emerged from longer positive responses, which accumulated during exploitative action-reward bouts, and hysteretic dynamics. Conversely, noradrenergic neurons in LC showed sustained inhibition of basal activity due to the accumulation of longer negative responses in LC. Chemogenetic manipulation of these sustained dynamics revealed that dopaminergic activity mediates action drive, whereas noradrenergic activity modulates choice diversity. These data uncover the emergence of sustained neural states in dopaminergic and noradrenergic networks that mediate dissociable aspects of exploitative bouts.
Collapse
Affiliation(s)
- Aaron C Koralek
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
- Champalimaud Neuroscience Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Rui M Costa
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA.
- Champalimaud Neuroscience Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| |
Collapse
|
177
|
Wu WL, Adame MD, Liou CW, Barlow JT, Lai TT, Sharon G, Schretter CE, Needham BD, Wang MI, Tang W, Ousey J, Lin YY, Yao TH, Abdel-Haq R, Beadle K, Gradinaru V, Ismagilov RF, Mazmanian SK. Microbiota regulate social behaviour via stress response neurons in the brain. Nature 2021; 595:409-414. [PMID: 34194038 DOI: 10.1038/s41586-021-03669-y] [Citation(s) in RCA: 184] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 05/25/2021] [Indexed: 02/07/2023]
Abstract
Social interactions among animals mediate essential behaviours, including mating, nurturing, and defence1,2. The gut microbiota contribute to social activity in mice3,4, but the gut-brain connections that regulate this complex behaviour and its underlying neural basis are unclear5,6. Here we show that the microbiome modulates neuronal activity in specific brain regions of male mice to regulate canonical stress responses and social behaviours. Social deviation in germ-free and antibiotic-treated mice is associated with elevated levels of the stress hormone corticosterone, which is primarily produced by activation of the hypothalamus-pituitary-adrenal (HPA) axis. Adrenalectomy, antagonism of glucocorticoid receptors, or pharmacological inhibition of corticosterone synthesis effectively corrects social deficits following microbiome depletion. Genetic ablation of glucocorticoid receptors in specific brain regions or chemogenetic inactivation of neurons in the paraventricular nucleus of the hypothalamus that produce corticotrophin-releasing hormone (CRH) reverse social impairments in antibiotic-treated mice. Conversely, specific activation of CRH-expressing neurons in the paraventricular nucleus induces social deficits in mice with a normal microbiome. Via microbiome profiling and in vivo selection, we identify a bacterial species, Enterococcus faecalis, that promotes social activity and reduces corticosterone levels in mice following social stress. These studies suggest that specific gut bacteria can restrain the activation of the HPA axis, and show that the microbiome can affect social behaviours through discrete neuronal circuits that mediate stress responses in the brain.
Collapse
Affiliation(s)
- Wei-Li Wu
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA. .,Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Mark D Adame
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Chia-Wei Liou
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jacob T Barlow
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Tzu-Ting Lai
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Gil Sharon
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Catherine E Schretter
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Brittany D Needham
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Madelyn I Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Weiyi Tang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - James Ousey
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Yuan-Yuan Lin
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Hsuan Yao
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Reem Abdel-Haq
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Keith Beadle
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Rustem F Ismagilov
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.,Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Sarkis K Mazmanian
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| |
Collapse
|
178
|
Vucic S, Pavey N, Haidar M, Turner BJ, Kiernan MC. Cortical hyperexcitability: Diagnostic and pathogenic biomarker of ALS. Neurosci Lett 2021; 759:136039. [PMID: 34118310 DOI: 10.1016/j.neulet.2021.136039] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 03/04/2021] [Accepted: 06/01/2021] [Indexed: 02/06/2023]
Abstract
Cortical hyperexcitability is an early and intrinsic feature of both sporadic and familial forms of amyotrophic lateral sclerosis (ALS).. Importantly, cortical hyperexcitability appears to be associated with motor neuron degeneration, possibly via an anterograde glutamate-mediated excitotoxic process, thereby forming a pathogenic basis for ALS. The presence of cortical hyperexcitability in ALS patients may be readily determined by transcranial magnetic stimulation (TMS), a neurophysiological tool that provides a non-invasive and painless method for assessing cortical function. Utilising the threshold tracking TMS technique, cortical hyperexcitability has been established as a robust diagnostic biomarker that distinguished ALS from mimicking disorders at early stages of the disease process. The present review discusses the pathophysiological and diagnostic utility of cortical hyperexcitability in ALS.
Collapse
Affiliation(s)
- Steve Vucic
- Western Clinical School, University of Sydney, Sydney, Australia.
| | - Nathan Pavey
- Western Clinical School, University of Sydney, Sydney, Australia
| | - Mouna Haidar
- Florey Institute of Neuroscieace and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Bradley J Turner
- Florey Institute of Neuroscieace and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre, University of Sydney and Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney, Australia
| |
Collapse
|
179
|
Pickering CA, Mazarakis ND. Viral Vector Delivery of DREADDs for CNS Therapy. Curr Gene Ther 2021; 21:191-206. [PMID: 33573551 DOI: 10.2174/1566523221666210211102435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/15/2021] [Accepted: 01/25/2021] [Indexed: 11/22/2022]
Abstract
Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) are genetically modified G-protein-coupled receptors (GPCRs), that can be activated by a synthetic ligand which is otherwise inert at endogenous receptors. DREADDs can be expressed in cells in the central nervous system (CNS) and subsequently offer the opportunity for remote and reversible silencing or activation of the target cells when the synthetic ligand is systemically administered. In neuroscience, DREADDs have thus far shown to be useful tools for several areas of research and offer considerable potential for the development of gene therapy strategies for neurological disorders. However, in order to design a DREADD-based gene therapy, it is necessary to first evaluate the viral vector delivery methods utilised in the literature to deliver these chemogenetic tools. This review evaluates each of the prominent strategies currently utilised for DREADD delivery, discussing their respective advantages and limitations. We focus on adeno-associated virus (AAV)-based and lentivirus-based systems, and the manipulation of these through cell-type specific promoters and pseudotyping. Furthermore, we address how virally mediated DREADD delivery could be improved in order to make it a viable gene therapy strategy and thus expand its translational potential.
Collapse
Affiliation(s)
- Ceri A Pickering
- Division of Neuroscience, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Nicholas D Mazarakis
- Division of Neuroscience, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
180
|
Ozawa A, Arakawa H. Chemogenetics drives paradigm change in the investigation of behavioral circuits and neural mechanisms underlying drug action. Behav Brain Res 2021; 406:113234. [PMID: 33741409 PMCID: PMC8110310 DOI: 10.1016/j.bbr.2021.113234] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/06/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022]
Abstract
Recent developments in chemogenetic approaches to the investigation of brain function have ushered in a paradigm change in the strategy for drug and behavior research and clinical drug-based medications. As the nature of the drug action is based on humoral regulation, it is a challenge to identify the neuronal mechanisms responsible for the expression of certain targeted behavior induced by drug application. The development of chemogenetic approaches has allowed researchers to control neural activities in targeted neurons through a toolbox, including engineered G protein-coupled receptors or ligand-gated ion channels together with exogenously inert synthetic ligands. This review provides a brief overview of the chemogenetics toolbox with an emphasis on the DREADDs (Designer Receptors Exclusively Activated by Designer Drugs) technique used in rodent models, which is applicable to the investigation of how specific neural circuits regulate behavioral processes. The use of chemogenetics has had a significant impact on basic neuroscience for a better understanding of the relationships between brain activity and the expression of behaviors with cell- and circuit-specific orders. Furthermore, chemogenetics is potentially a useful tool to deconstruct the neuropathological mechanisms of mental diseases and its regulation by drug, and provide us with transformative therapeutics with medication. We also review recent findings in the use of chemogenetic techniques to uncover functional circuit connections of serotonergic neurons in rodent models.
Collapse
Affiliation(s)
- Akihiko Ozawa
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL, USA
| | - Hiroyuki Arakawa
- Department of Psychology, Tokiwa University, Mito, Ibaraki, Japan; Department of Systems Physiology, University of Ryukyus, Faculty of Medicine, Nakagami District, Okinawa, Japan.
| |
Collapse
|
181
|
Taylor-Yeremeeva EM, Wisser SC, Chakoma TL, Aldrich SJ, Denney AE, Donahue EK, Adelman JS, Ihle PCJ, Robinson S. Appetitive and aversive sensory preconditioning in rats is impaired by disruption of the postrhinal cortex. Neurobiol Learn Mem 2021; 183:107461. [PMID: 34015445 DOI: 10.1016/j.nlm.2021.107461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 04/20/2021] [Accepted: 05/12/2021] [Indexed: 11/25/2022]
Abstract
Episodic memory involves binding stimuli and/or events together in time and place. Furthermore, memories become more complex when new experiences influence the meaning of stimuli within the original memory. Thus collectively, complex episodic memory formation and maintenance involves processes such as encoding, storage, retrieval, updating and reconsolidation, which can be studied using animal models of higher-order conditioning. In the present study aversive and appetitive sensory preconditioning paradigms were used to test the hypothesis that the postrhinal cortex (POR), which is a component of the hippocampal memory system, is involved in higher-order conditioning. Drawing on the known role of the POR in contextual learning, Experiment 1 employed a four-phase sensory preconditioning task that involved fear learning and context discrimination in rats with or without permanent lesions of the POR. In parallel, to examine POR function during higher-order conditioning in the absence of a particular spatial arrangement, Experiments 2 and 3 used a three-phase sensory preconditioning paradigm involving phasic stimuli. In Experiment 2, bilateral lesions of the POR were made and in Experiment 3, a chemogenetic approach was used to temporarily inactivate POR neurons during each phase of the paradigm. Evidence of successful sensory preconditioning was observed in sham rats which, during the critical context discrimination test, demonstrated higher levels of freezing behavior when re-exposed to the paired versus the unpaired context, whereas POR-lesioned rats did not. Data from the appetitive sensory preconditioning paradigm also confirmed the hypothesis in that during the critical auditory discrimination test, sham rats showed greater food cup responding following presentations of the paired compared to the unpaired auditory stimulus, whereas POR-lesioned rats did not. Lastly, in Experiment 3, when the POR was inactivated only during preconditioning or only during conditioning, discrimination during the critical auditory test was impaired. Thus, regardless of whether stimulus-stimulus associations were formed between static or phasic stimuli or whether revaluation of the paired stimulus occurred through association with an aversive or an appetitive unconditioned stimulus, the effects were the same; POR lesions disrupted the ability to use higher-order conditioned stimuli to guide prospective behavior.
Collapse
Affiliation(s)
| | - Stephen C Wisser
- Program in Neuroscience, Hamilton College, Clinton, NY 13323, USA
| | | | - Sara J Aldrich
- Program in Neuroscience, Hamilton College, Clinton, NY 13323, USA
| | - Amelia E Denney
- Program in Neuroscience, Hamilton College, Clinton, NY 13323, USA
| | - Erin K Donahue
- Program in Neuroscience, Hamilton College, Clinton, NY 13323, USA
| | - Julia S Adelman
- Department of Neuroscience, Oberlin College, Oberlin, OH 44074, USA
| | - Peter C J Ihle
- Department of Neuroscience, Oberlin College, Oberlin, OH 44074, USA
| | - Siobhan Robinson
- Program in Neuroscience, Hamilton College, Clinton, NY 13323, USA; Department of Psychology, Hamilton College, Clinton, NY 13323, USA.
| |
Collapse
|
182
|
Shapiro LP, Pitts EG, Li DC, Barbee BR, Hinton EA, Bassell GJ, Gross C, Gourley SL. The PI3-Kinase p110β Isoform Controls Severity of Cocaine-Induced Sequelae and Alters the Striatal Transcriptome. Biol Psychiatry 2021; 89:959-969. [PMID: 33773752 PMCID: PMC8202243 DOI: 10.1016/j.biopsych.2021.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/04/2021] [Accepted: 01/13/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND The PI3-kinase (PI3K) complex is a well-validated target for mitigating cocaine-elicited sequelae, but pan-PI3K inhibitors are not viable long-term treatment options. The PI3K complex is composed of p110 catalytic and regulatory subunits, which can be individually manipulated for therapeutic purposes. However, this possibility has largely not been explored in behavioral contexts. METHODS Here, we inhibited PI3K p110β in the medial prefrontal cortex (mPFC) of cocaine-exposed mice. Behavioral models for studying relapse, sensitization, and decision-making biases were paired with protein quantification, RNA sequencing, and cell type-specific chemogenetic manipulation and RNA quantification to determine whether and how inhibiting PI3K p110β confers resilience to cocaine. RESULTS Viral-mediated PI3K p110β silencing reduced cue-induced reinstatement of cocaine seeking by half, blocked locomotor sensitization, and restored mPFC synaptic marker content after exposure to cocaine. Cocaine blocked the ability of mice to select actions based on their consequences, and p110β inhibition restored this ability. Silencing dopamine D2 receptor-expressing excitatory mPFC neurons mimicked cocaine, impairing goal-seeking behavior, and again, p110β inhibition restored goal-oriented action. We verified the presence of p110β in mPFC neurons projecting to the dorsal striatum and orbitofrontal cortex and found that inhibiting p110β in the mPFC altered the expression of functionally defined gene clusters within the dorsal striatum and not orbitofrontal cortex. CONCLUSIONS Subunit-selective PI3K silencing potently mitigates drug seeking, sensitization, and decision-making biases after exposure to cocaine. We suggest that inhibiting PI3K p110β provides neuroprotection against cocaine by triggering coordinated corticostriatal adaptations.
Collapse
Affiliation(s)
- Lauren P. Shapiro
- Graduate Program in Molecular and Systems Pharmacology, Emory University,Department of Pediatrics, Emory University School of Medicine; Yerkes National Primate Research Center
| | - Elizabeth G. Pitts
- Department of Pediatrics, Emory University School of Medicine; Yerkes National Primate Research Center,Graduate Program in Neuroscience, Emory University
| | - Dan C. Li
- Department of Pediatrics, Emory University School of Medicine; Yerkes National Primate Research Center,Graduate Program in Neuroscience, Emory University
| | - Britton R. Barbee
- Graduate Program in Molecular and Systems Pharmacology, Emory University,Department of Pediatrics, Emory University School of Medicine; Yerkes National Primate Research Center
| | - Elizabeth A. Hinton
- Department of Pediatrics, Emory University School of Medicine; Yerkes National Primate Research Center,Graduate Program in Neuroscience, Emory University
| | - Gary J. Bassell
- Graduate Program in Neuroscience, Emory University,Department of Cell Biology, Emory University
| | - Christina Gross
- Division of Neurology, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine
| | - Shannon L. Gourley
- Department of Pediatrics, Emory University School of Medicine; Yerkes National Primate Research Center,Graduate Program in Neuroscience, Emory University,Children’s Healthcare of Atlanta
| |
Collapse
|
183
|
Guo W, Wan X, Ma L, Zhang J, Hashimoto K. Abnormalities in the composition of the gut microbiota in mice after repeated administration of DREADD ligands. Brain Res Bull 2021; 173:66-73. [PMID: 34004259 DOI: 10.1016/j.brainresbull.2021.05.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/06/2021] [Accepted: 05/13/2021] [Indexed: 02/08/2023]
Abstract
Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) are known as genetically modified G-protein-coupled receptors (GPCRs), which can be activated by synthetic ligands such as clozapine N-oxide (CNO) and DREADD agonist 21 (compound 21: C21). The brain-gut-microbiota axis has a crucial role in bidirectional interactions between the brain and the gastrointestinal microbiota. In this study, we investigated whether repeated administration of CNO or C21 could influence the gut microbiota and short-chain fatty acids (SCFAs) in feces of adult mice. Repeated administration of CNO or C21 as drinking water did not alter the α- and β-diversity of gut microbiota in mice compared with control mice. However, we found significant changes in relative abundance for several bacteria in the CNO (or C21) group at the taxonomic level compared to the control group. The linear discriminant analysis effect size (LEfSe) algorithm distinguished the family Prevotellaceae, the genus Anaerocolumna, the genus Prevotella, and the genus Frisingicoccus, these four specific microbial markers for the CNO group relative to the control group. In addition, the LEfSe algorithm identified the family Clostridiaceae, the genus Faecalicatena and the genus Marinisporobacter, these three bacteria of different taxonomic as potential microbial markers for the C21 group relative to the control group. In contrast, repeated administration of CNO (or C21) did not alter SCFAs in feces samples of adult mice. The data suggest that repeated administration of CNO or C21 contributes to an unusual organization of the gut microbiota in adult mice. Therefore, abnormalities in the composition of gut microbiota by repeated dosing of DREADD ligands should be taken into consideration for behavioral and biological functions in rodents treated with DREADD ligands.
Collapse
Affiliation(s)
- Wei Guo
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Xiayun Wan
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Li Ma
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Jiancheng Zhang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan.
| |
Collapse
|
184
|
Social isolation uncovers a circuit underlying context-dependent territory-covering micturition. Proc Natl Acad Sci U S A 2021; 118:2018078118. [PMID: 33443190 DOI: 10.1073/pnas.2018078118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The release of urine, or micturition, serves a fundamental physiological function and, in many species, is critical for social communication. In mice, the pattern of urine release is modulated by external and internal factors and transmitted to the spinal cord via the pontine micturition center (PMC). Here, we exploited a behavioral paradigm in which mice, depending on strain, social experience, and sensory context, either vigorously cover an arena with small urine spots or deposit urine in a few isolated large spots. We refer to these micturition modes as, respectively, high and low territory-covering micturition (TCM) and find that the presence of a urine stimulus robustly induces high TCM in socially isolated mice. Comparison of the brain networks activated by social isolation and by urine stimuli to those upstream of the PMC identified the lateral hypothalamic area as a potential modulator of micturition modes. Indeed, chemogenetic manipulations of the lateral hypothalamus can switch micturition behavior between high and low TCM, overriding the influence of social experience and sensory context. Our results suggest that both inhibitory and excitatory signals arising from a network upstream of the PMC are integrated to determine context- and social-experience-dependent micturition patterns.
Collapse
|
185
|
Rodgers J, Bano‐Otalora B, Belle MDC, Paul S, Hughes R, Wright P, McDowell R, Milosavljevic N, Orlowska‐Feuer P, Martial FP, Wynne J, Ballister ER, Storchi R, Allen AE, Brown T, Lucas RJ. Using a bistable animal opsin for switchable and scalable optogenetic inhibition of neurons. EMBO Rep 2021; 22:e51866. [PMID: 33655694 PMCID: PMC8097317 DOI: 10.15252/embr.202051866] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 11/09/2022] Open
Abstract
There is no consensus on the best inhibitory optogenetic tool. Since Gi/o signalling is a native mechanism of neuronal inhibition, we asked whether Lamprey Parapinopsin ("Lamplight"), a Gi/o-coupled bistable animal opsin, could be used for optogenetic silencing. We show that short (405 nm) and long (525 nm) wavelength pulses repeatedly switch Lamplight between stable signalling active and inactive states, respectively, and that combining these wavelengths can be used to achieve intermediate levels of activity. These properties can be applied to produce switchable neuronal hyperpolarisation and suppression of spontaneous spike firing in the mouse hypothalamic suprachiasmatic nucleus. Expressing Lamplight in (predominantly) ON bipolar cells can photosensitise retinas following advanced photoreceptor degeneration, with 405 and 525 nm stimuli producing responses of opposite sign in the output neurons of the retina. We conclude that bistable animal opsins can co-opt endogenous signalling mechanisms to allow optogenetic inhibition that is scalable, sustained and reversible.
Collapse
Affiliation(s)
- Jessica Rodgers
- Faculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
| | | | - Mino D C Belle
- Institute of Biomedical and Clinical SciencesUniversity of Exeter Medical SchoolUniversity of ExeterExeterUK
| | - Sarika Paul
- Faculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
| | - Rebecca Hughes
- Faculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
| | - Phillip Wright
- Faculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
| | - Richard McDowell
- Faculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
| | - Nina Milosavljevic
- Faculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
| | - Patrycja Orlowska‐Feuer
- Faculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
- Department of Neurophysiology and ChronobiologyInstitute of Zoology and Biomedical ResearchJagiellonian University in KrakowKrakowPoland
| | - Franck P Martial
- Faculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
| | - Jonathan Wynne
- Faculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
| | - Edward R Ballister
- Faculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
- Department of Biomedical EngineeringColumbia UniversityNew YorkNYUSA
| | - Riccardo Storchi
- Faculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
| | - Annette E Allen
- Faculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
| | - Timothy Brown
- Faculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
| | - Robert J Lucas
- Faculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
| |
Collapse
|
186
|
Lattanzi W, Ripoli C, Greco V, Barba M, Iavarone F, Minucci A, Urbani A, Grassi C, Parolini O. Basic and Preclinical Research for Personalized Medicine. J Pers Med 2021; 11:jpm11050354. [PMID: 33946634 PMCID: PMC8146055 DOI: 10.3390/jpm11050354] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/19/2021] [Accepted: 04/26/2021] [Indexed: 12/18/2022] Open
Abstract
Basic and preclinical research founded the progress of personalized medicine by providing a prodigious amount of integrated profiling data and by enabling the development of biomedical applications to be implemented in patient-centered care and cures. If the rapid development of genomics research boosted the birth of personalized medicine, further development in omics technologies has more recently improved our understanding of the functional genome and its relevance in profiling patients’ phenotypes and disorders. Concurrently, the rapid biotechnological advancement in diverse research areas enabled uncovering disease mechanisms and prompted the design of innovative biological treatments tailored to individual patient genotypes and phenotypes. Research in stem cells enabled clarifying their role in tissue degeneration and disease pathogenesis while providing novel tools toward the development of personalized regenerative medicine strategies. Meanwhile, the evolving field of integrated omics technologies ensured translating structural genomics information into actionable knowledge to trace detailed patients’ molecular signatures. Finally, neuroscience research provided invaluable models to identify preclinical stages of brain diseases. This review aims at discussing relevant milestones in the scientific progress of basic and preclinical research areas that have considerably contributed to the personalized medicine revolution by bridging the bench-to-bed gap, focusing on stem cells, omics technologies, and neuroscience fields as paradigms.
Collapse
Affiliation(s)
- Wanda Lattanzi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (W.L.); (C.R.); (V.G.); (M.B.); (F.I.); (A.M.); (A.U.); (C.G.)
- Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Cristian Ripoli
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (W.L.); (C.R.); (V.G.); (M.B.); (F.I.); (A.M.); (A.U.); (C.G.)
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Viviana Greco
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (W.L.); (C.R.); (V.G.); (M.B.); (F.I.); (A.M.); (A.U.); (C.G.)
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marta Barba
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (W.L.); (C.R.); (V.G.); (M.B.); (F.I.); (A.M.); (A.U.); (C.G.)
- Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Federica Iavarone
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (W.L.); (C.R.); (V.G.); (M.B.); (F.I.); (A.M.); (A.U.); (C.G.)
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Angelo Minucci
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (W.L.); (C.R.); (V.G.); (M.B.); (F.I.); (A.M.); (A.U.); (C.G.)
| | - Andrea Urbani
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (W.L.); (C.R.); (V.G.); (M.B.); (F.I.); (A.M.); (A.U.); (C.G.)
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Claudio Grassi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (W.L.); (C.R.); (V.G.); (M.B.); (F.I.); (A.M.); (A.U.); (C.G.)
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Ornella Parolini
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (W.L.); (C.R.); (V.G.); (M.B.); (F.I.); (A.M.); (A.U.); (C.G.)
- Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Correspondence:
| |
Collapse
|
187
|
Poth KM, Texakalidis P, Boulis NM. Chemogenetics: Beyond Lesions and Electrodes. Neurosurgery 2021; 89:185-195. [PMID: 33913505 PMCID: PMC8279839 DOI: 10.1093/neuros/nyab147] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 02/26/2021] [Indexed: 01/14/2023] Open
Abstract
The field of chemogenetics has rapidly expanded over the last decade, and engineered receptors are currently utilized in the lab to better understand molecular interactions in the nervous system. We propose that chemogenetic receptors can be used for far more than investigational purposes. The potential benefit of adding chemogenetic neuromodulation to the current neurosurgical toolkit is substantial. There are several conditions currently treated surgically, electrically, and pharmacologically in clinic, and this review highlights how chemogenetic neuromodulation could improve patient outcomes over current neurosurgical techniques. We aim to emphasize the need to take these techniques from bench to bedside.
Collapse
Affiliation(s)
- Kelly M Poth
- Department of Neurosurgery, Emory University, Atlanta, Georgia, USA
| | | | | |
Collapse
|
188
|
Chemogenetic Activation of CX3CR1-Expressing Spinal Microglia Using Gq-DREADD Elicits Mechanical Allodynia in Male Mice. Cells 2021; 10:cells10040874. [PMID: 33921365 PMCID: PMC8069983 DOI: 10.3390/cells10040874] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/05/2021] [Accepted: 04/09/2021] [Indexed: 12/11/2022] Open
Abstract
It is important to investigate the sex-dependent roles of microglia in pain hypersensitivity as reactive microglia within the spinal dorsal horn (DH) have been reported to be pivotal in neuropathic pain induction in male rodents upon nerve injury. Here, we aimed at determining the role of sex differences in the behavioral and functional outcomes of the chemogenetic activation of spinal microglia using Gq-designer receptors exclusively activated by designer drugs (Gq-DREADD) driven by the microglia-specific Cx3cr1 promoter. CAG-LSL-human Gq-coupled M3 muscarinic receptors (hM3Dq)-DREADD mice were crossed with CX3C chemokine receptor 1 (CX3CR1)-Cre mice, and immunohistochemistry images revealed that hM3Dq was selectively expressed on Iba1+ microglia, but not on astrocytes and neurons. Intrathecal (i.t.) administration of clozapine-N-oxide (CNO) elicited mechanical allodynia exclusively in male mice. Furthermore, the reactive microglia-dominant molecules that contributed to pain hypersensitivity in CX3CR1-hM3Dq were upregulated in mice of both sexes. The degree of upregulation was greater in male than in female mice. Depletion of spinal microglia using pexidartinib (PLX3397), a colony stimulating factor-1 receptor inhibitor, alleviated the male CX3CR1-hM3Dq mice from pain hypersensitivity and compromised the expression of inflammatory molecules. Thus, the chemogenetic activation of spinal microglia resulted in pain hypersensitivity in male mice, suggesting the sex-dependent molecular aspects of spinal microglia in the regulation of pain.
Collapse
|
189
|
Ma N, Nivedha AK, Vaidehi N. Allosteric communication regulates ligand-specific GPCR activity. FEBS J 2021; 288:2502-2512. [PMID: 33738925 PMCID: PMC9805801 DOI: 10.1111/febs.15826] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 01/11/2023]
Abstract
G protein-coupled receptors (GPCRs) are membrane-bound proteins that are ubiquitously expressed in many cell types and take part in mediating multiple signaling pathways. GPCRs are dynamic proteins and exist in an equilibrium between an ensemble of conformational states such as inactive and fully active states. This dynamic nature of GPCRs is one of the factors that confers their basal activity even in the absence of any ligand-mediated activation. Ligands selectively bind and stabilize a subset of the conformations from the ensemble leading to a shift in the equilibrium toward the inactive or the active state depending on the nature of the ligand. This ligand-selective effect is achieved through allosteric communication between the ligand binding site and G protein or β-arrestin coupling site. Similarly, the G protein coupling to the receptor exerts the allosteric effect on the ligand binding region leading to increased binding affinity for agonists and decreased affinity for antagonists or inverse agonists. In this review, we enumerate the current state of our understanding of the mechanism of allosteric communication in GPCRs with a specific focus on the critical role of computational methods in delineating the residues involved in allosteric communication. Analyzing allosteric communication mechanism using molecular dynamics simulations has revealed (a) a structurally conserved mechanism of allosteric communication that regulates the G protein coupling, (b) a rational structure-based approach to designing selective ligands, and (c) an approach to designing allosteric GPCR mutants that are either ligand and G protein or β-arrestin selective.
Collapse
Affiliation(s)
- Ning Ma
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA 91010
| | - Anita K. Nivedha
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA 91010
| | - Nagarajan Vaidehi
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA 91010
| |
Collapse
|
190
|
Rouleau N, Murugan NJ, Kaplan DL. Toward Studying Cognition in a Dish. Trends Cogn Sci 2021; 25:294-304. [PMID: 33546973 PMCID: PMC7946736 DOI: 10.1016/j.tics.2021.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 12/31/2022]
Abstract
Bioengineered neural tissues help advance our understanding of neurodevelopment, regeneration, and neural disease; however, it remains unclear whether they can replicate higher-order functions including cognition. Building upon technical achievements in the fields of biomaterials, tissue engineering, and cell biology, investigators have generated an assortment of artificial brain structures and cocultured circuits. Though they have displayed basic electrochemical signaling, their capacities to generate minimal patterns of information processing suggestive of high-order cognitive analogues have not yet been explored. Here, we review the current state of neural tissue engineering and consider the possibility of a study of cognition in vitro. We adopt a practical definition of minimal cognition, anticipate problems of measurement, and discuss solutions toward a study of cognition in a dish.
Collapse
Affiliation(s)
- Nicolas Rouleau
- Department of Psychology, Algoma University, 1520 Queen Street East, Sault Ste. Marie, Ontario, Canada, P6A 2G4; Department of Biomedical Engineering, Tufts University, Science and Technology Center, 4 Colby Street, Medford, MA 02155, USA
| | - Nirosha J Murugan
- Department of Biology, Algoma University, 1520 Queen Street East, Sault Ste. Marie, Ontario, Canada, P6A 2G4
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Science and Technology Center, 4 Colby Street, Medford, MA 02155, USA.
| |
Collapse
|
191
|
Huang Q, Garrett A, Bose S, Blocker S, Rios AC, Clevers H, Shen X. The frontier of live tissue imaging across space and time. Cell Stem Cell 2021; 28:603-622. [PMID: 33798422 PMCID: PMC8034393 DOI: 10.1016/j.stem.2021.02.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
What you see is what you get-imaging techniques have long been essential for visualization and understanding of tissue development, homeostasis, and regeneration, which are driven by stem cell self-renewal and differentiation. Advances in molecular and tissue modeling techniques in the last decade are providing new imaging modalities to explore tissue heterogeneity and plasticity. Here we describe current state-of-the-art imaging modalities for tissue research at multiple scales, with a focus on explaining key tradeoffs such as spatial resolution, penetration depth, capture time/frequency, and moieties. We explore emerging tissue modeling and molecular tools that improve resolution, specificity, and throughput.
Collapse
Affiliation(s)
- Qiang Huang
- Department of Pediatric Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004 Shaanxi, China; Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Aliesha Garrett
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Shree Bose
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Stephanie Blocker
- Center for In Vitro Microscopy, Duke University, Durham, NC 27708, USA
| | - Anne C Rios
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584, the Netherlands; Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht 3584, the Netherlands
| | - Hans Clevers
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584, the Netherlands; Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht 3584, the Netherlands; Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center (UMC) Utrecht, Utrecht 3584, the Netherlands
| | - Xiling Shen
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
192
|
Chadney OMT, Blankvoort S, Grimstvedt JS, Utz A, Kentros CG. Multiplexing viral approaches to the study of the neuronal circuits. J Neurosci Methods 2021; 357:109142. [PMID: 33753126 DOI: 10.1016/j.jneumeth.2021.109142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 02/27/2021] [Accepted: 03/10/2021] [Indexed: 12/16/2022]
Abstract
Neural circuits are composed of multitudes of elaborately interconnected cell types. Understanding neural circuit function requires not only cell-specific knowledge of connectivity, but the ability to record and manipulate distinct cell types independently. Recent advances in viral vectors promise the requisite specificity to perform true "circuit-breaking" experiments. However, such new avenues of multiplexed, cell-specific investigation raise new technical issues: one must ensure that both the viral vectors and their transgene payloads do not overlap with each other in both an anatomical and a functional sense. This review describes benefits and issues regarding the use of viral vectors to analyse the function of neural circuits and provides a resource for the design and implementation of such multiplexing experiments.
Collapse
Affiliation(s)
- Oscar M T Chadney
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim, Norway.
| | - Stefan Blankvoort
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim, Norway
| | - Joachim S Grimstvedt
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim, Norway
| | - Annika Utz
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim, Norway
| | - Clifford G Kentros
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim, Norway.
| |
Collapse
|
193
|
Abreu N, Levitz J. Optogenetic Techniques for Manipulating and Sensing G Protein-Coupled Receptor Signaling. Methods Mol Biol 2021; 2173:21-51. [PMID: 32651908 DOI: 10.1007/978-1-0716-0755-8_2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
G protein-coupled receptors (GPCRs) form the largest class of membrane receptors in the mammalian genome with nearly 800 human genes encoding for unique subtypes. Accordingly, GPCR signaling is implicated in nearly all physiological processes. However, GPCRs have been difficult to study due in part to the complexity of their function which can lead to a plethora of converging or diverging downstream effects over different time and length scales. Classic techniques such as pharmacological control, genetic knockout and biochemical assays often lack the precision required to probe the functions of specific GPCR subtypes. Here we describe the rapidly growing set of optogenetic tools, ranging from methods for optical control of the receptor itself to optical sensing and manipulation of downstream effectors. These tools permit the quantitative measurements of GPCRs and their downstream signaling with high specificity and spatiotemporal precision.
Collapse
Affiliation(s)
- Nohely Abreu
- Biochemistry, Cell and Molecular Biology Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Joshua Levitz
- Biochemistry, Cell and Molecular Biology Graduate Program, Weill Cornell Medicine, New York, NY, USA.
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
194
|
Phillips JA, Hutchings C, Djamgoz MBA. Clinical Potential of Nerve Input to Tumors: A Bioelectricity Perspective. Bioelectricity 2021; 3:14-26. [PMID: 34476375 DOI: 10.1089/bioe.2020.0051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
We support the notion that the neural connections of the tumor microenvironment (TME) and the associated 'bioelectricity' play significant role in the pathophysiology of cancer. In several cancers, the nerve input promotes the cancer process. While straightforward surgical denervation of tumors, therefore, could improve prognosis, resulting side effects of such a procedure would be unpredictable and irreversible. On the other hand, tumor innervation can be manipulated effectively for therapeutic purposes by alternative novel approaches broadly termed "electroceuticals." In this perspective, we evaluate the clinical potential of targeting the TME first through manipulation of the nerve input itself and second by application of electric fields directly to the tumor. The former encompasses several different biophysical and biochemical approaches. These include implantable devices, nanoparticles, and electroactive polymers, as well as optogenetics and chemogenetics. As regard bioelectrical manipulation of the tumor itself, the "tumor-treating field" technique, applied to gliomas commonly in combination with chemotherapy, is evaluated. Also, as electroceuticals, drugs acting on ion channels and neurotransmitter receptors are highlighted for completeness. It is concluded, first, that electroceuticals comprise a broad range of biomedical tools. Second, such electroceuticals present significant clinical potential for exploiting the neural component of the TME as a strategy against cancer. Finally, the inherent bioelectric characteristics of tumors themselves are also amenable to complementary approaches. Collectively, these represent an evolving, dynamic field and further progress and applications can be expected to follow both conceptually and technically.
Collapse
Affiliation(s)
- Jade A Phillips
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Charlotte Hutchings
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, London, United Kingdom.,Biotechnology Research Center, Cyprus International University, Nicosia, North Cyprus
| |
Collapse
|
195
|
Dieterich A, Floeder J, Stech K, Lee J, Srivastava P, Barker DJ, Samuels BA. Activation of Basolateral Amygdala to Nucleus Accumbens Projection Neurons Attenuates Chronic Corticosterone-Induced Behavioral Deficits in Male Mice. Front Behav Neurosci 2021; 15:643272. [PMID: 33716685 PMCID: PMC7943928 DOI: 10.3389/fnbeh.2021.643272] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/28/2021] [Indexed: 11/13/2022] Open
Abstract
The basolateral amygdala (BLA) is critical for reward behaviors via a projection to the nucleus accumbens (NAc). Specifically, BLA-NAc projections are involved in reinforcement learning, reward-seeking, sustained instrumental responding, and risk behaviors. However, it remains unclear whether chronic stress interacts with BLA-NAc projection neurons to result in maladaptive behaviors. Here we take a chemogenetic, projection-specific approach to clarify how NAc-projecting BLA neurons affect avoidance, reward, and feeding behaviors in male mice. Then, we examine whether chemogenetic activation of NAc-projecting BLA neurons attenuates the maladaptive effects of chronic corticosterone (CORT) administration on these behaviors. CORT mimics the behavioral and neural effects of chronic stress exposure. We found a nuanced role of BLA-NAc neurons in mediating reward behaviors. Surprisingly, activation of BLA-NAc projections rescues CORT-induced deficits in the novelty suppressed feeding, a behavior typically associated with avoidance. Activation of BLA-NAc neurons also increases instrumental reward-seeking without affecting free-feeding in chronic CORT mice. Taken together, these data suggest that NAc-projecting BLA neurons are involved in chronic CORT-induced maladaptive reward and motivation behaviors.
Collapse
Affiliation(s)
- Andrew Dieterich
- Neuroscience Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
- Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Joseph Floeder
- Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Karina Stech
- Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Jay Lee
- Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Prachi Srivastava
- Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - David J. Barker
- Neuroscience Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
- Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Benjamin A. Samuels
- Neuroscience Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
- Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| |
Collapse
|
196
|
Koyanagi M, Saito T, Wada S, Nagata T, Kawano-Yamashita E, Terakita A. Optogenetic Potentials of Diverse Animal Opsins: Parapinopsin, Peropsin, LWS Bistable Opsin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1293:141-151. [PMID: 33398811 DOI: 10.1007/978-981-15-8763-4_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Animal opsin-based pigments are light-activated G-protein-coupled receptors (GPCRs), which drive signal transduction cascades via G-proteins. Thousands of animal opsins have been identified, and molecular phylogenetic and biochemical analyses have revealed the unexpected diversity in selectivity of G-protein activation and photochemical property. Here we discuss the optogenetic potentials of diverse animal opsins, particularly recently well-characterized three non-canonical opsins, parapinopsin, peropsin, and LWS bistable opsin. Unlike canonical opsins such as vertebrate visual opsins that have been conventionally used for optogenetic applications, these opsins are bistable; opsin-based pigments do not release the chromophore retinal after light absorption, and the stable photoproducts revert to their original dark states upon subsequent light absorption. Parapinopsins have a "complete photoregeneration ability," which allows a clear color-dependent regulation of signal transductions. On the other hand, peropsins serve as a "dark-active and light-inactivated" GPCR to regulate signal transductions in the opposite way compared with usual opsins. In addition, an LWS bistable opsin from a butterfly was revealed to be the longest wavelength-sensitive animal opsin with its absorption maximum at ~570 nm. The property-dependent optical regulations of signal transductions were demonstrated in mammalian cultured cells, showing potentials of new optogenetic tools.
Collapse
Affiliation(s)
- Mitsumasa Koyanagi
- Department of Biology and Geosciences, Graduate School of Science, Osaka City University, Osaka, Japan
| | - Tomoka Saito
- Department of Biology and Geosciences, Graduate School of Science, Osaka City University, Osaka, Japan
| | - Seiji Wada
- Department of Biology and Geosciences, Graduate School of Science, Osaka City University, Osaka, Japan
| | - Takashi Nagata
- Department of Biology and Geosciences, Graduate School of Science, Osaka City University, Osaka, Japan
| | - Emi Kawano-Yamashita
- Department of Biology and Geosciences, Graduate School of Science, Osaka City University, Osaka, Japan
| | - Akihisa Terakita
- Department of Biology and Geosciences, Graduate School of Science, Osaka City University, Osaka, Japan.
| |
Collapse
|
197
|
Chemogenetic manipulation of astrocytic activity: Is it possible to reveal the roles of astrocytes? Biochem Pharmacol 2021; 186:114457. [PMID: 33556341 DOI: 10.1016/j.bcp.2021.114457] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 01/08/2023]
Abstract
Astrocytes are the major glial cells in the central nervous system, but unlike neurons, they do not produce action potentials. For many years, astrocytes were considered supporting cells in the central nervous system (CNS). Technological advances over the last two decades are changing the face of glial research. Accumulating data from recent investigations show that astrocytes display transient calcium spikes and regulate synaptic transmission by releasing transmitters called gliotransmitters. Many new powerful technologies are used to interfere with astrocytic activity, in order to obtain a better understanding of the roles of astrocytes in the brain. Among these technologies, chemogenetics has recently been used frequently. In this review, we will summarize new functions of astrocytes in the brain that have been revealed using this cutting-edge technique. Moreover, we will discuss the possibilities and challenges of manipulating astrocytic activity using this technology.
Collapse
|
198
|
Yi MH, Liu YU, Liu K, Chen T, Bosco DB, Zheng J, Xie M, Zhou L, Qu W, Wu LJ. Chemogenetic manipulation of microglia inhibits neuroinflammation and neuropathic pain in mice. Brain Behav Immun 2021; 92:78-89. [PMID: 33221486 PMCID: PMC7897256 DOI: 10.1016/j.bbi.2020.11.030] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 12/21/2022] Open
Abstract
Microglia play an important role in the central sensitization and chronic pain. However, a direct connection between microglial function and pain development in vivo remains incompletely understood. To address this issue, we applied chemogenetic approach by using CX3CR1creER/+:R26LSL-hM4Di/+ transgenic mice to enable expression of inhibitory Designer Receptors Exclusively Activated by Designer Drugs (Gi DREADD) in microglia. We found that microglial Gi DREADD activation inhibited spinal nerve transection (SNT)-induced microglial reactivity as well as chronic pain in both male and female mice. Gi DREADD activation downregulated the transcription factor interferon regulatory factor 8 (IRF8) and its downstream target pro-inflammatory cytokine interleukin 1 beta (IL-1β). Using in vivo spinal cord recording, we found that activation of microglial Gi DREADD attenuated synaptic transmission following SNT. Our results demonstrate that microglial Gi DREADD reduces neuroinflammation, synaptic function and neuropathic pain after SNT. Thus, chemogenetic approaches provide a potential opportunity for interrogating microglial function and neuropathic pain treatment.
Collapse
Affiliation(s)
- Min-Hee Yi
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yong U. Liu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Kevin Liu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, 08854 USA
| | - Tingjun Chen
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Dale B. Bosco
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Manling Xie
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Lijun Zhou
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Wenchun Qu
- Department of Pain Medicine, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Departments of Immunology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
199
|
Maldonado R, Calvé P, García-Blanco A, Domingo-Rodriguez L, Senabre E, Martín-García E. Vulnerability to addiction. Neuropharmacology 2021; 186:108466. [PMID: 33482225 DOI: 10.1016/j.neuropharm.2021.108466] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/03/2020] [Accepted: 01/14/2021] [Indexed: 12/22/2022]
Abstract
Addiction is a chronic brain disease that has dramatic health and socioeconomic consequences worldwide. Multiple approaches have been used for decades to clarify the neurobiological basis of this disease and to identify novel potential treatments. This review summarizes the main brain networks involved in the vulnerability to addiction and specific innovative technological approaches to investigate these neural circuits. First, the evolution of the definition of addiction across the Diagnostic and Statistical Manual of Mental Disorders (DSM) is revised. We next discuss several innovative experimental techniques that, combined with behavioral approaches, have allowed recent critical advances in understanding the neural circuits involved in addiction, including DREADDs, calcium imaging, and electrophysiology. All these techniques have been used to investigate specific neural circuits involved in vulnerability to addiction and have been extremely useful to clarify the neurobiological basis of each specific component of the addictive process. These novel tools targeting specific brain regions are of great interest to further understand the different aspects of this complex disease. This article is part of the special issue on 'Vulnerabilities to Substance Abuse.'.
Collapse
Affiliation(s)
- R Maldonado
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain; Hospital Del Mar Medical Research Institute (IMIM), Barcelona, Spain.
| | - P Calvé
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - A García-Blanco
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - L Domingo-Rodriguez
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - E Senabre
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - E Martín-García
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| |
Collapse
|
200
|
Vincent SG, Fisher JT. In vivo cardiopulmonary impact of skeletal M 3Dq DREADD expression: a pilot study. J Comp Physiol B 2021; 191:1059-1070. [PMID: 34272586 PMCID: PMC8572194 DOI: 10.1007/s00360-021-01387-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 05/23/2021] [Accepted: 07/06/2021] [Indexed: 01/07/2023]
Abstract
The muscarinic M3 receptor (M3R) is implicated in cardiopulmonary control and many other peripheral physiologic functions. Previous observations report mortality in mice expressing a Gq-linked designer G-protein coupled receptor (Dq) selectively in striated muscle, while M3Dq DREADD (Designer Receptor Exclusively Activated by Designer Drug), selectively expressed in skeletal muscle (SKM) impacts glucose metabolism. We investigated whether activation of SKM M3Dq impacts cardiopulmonary function. Heart rate (HR), body temperature (Tb) and locomotor activity (ACT) were measured in 4 conscious, chronically instrumented M3Dq DREADD mice and 4 wildtype controls. Circadian values of HR, BT and ACT were not different between genotypes (p > 0.05). Activation of the M3Dq DREADD by clozapine N-oxide (CNO; 0.1 mg/kg) resulted in: a significant drop in heart rate, 2 h after injection, compared with a time-matched baseline control period from the same animals (460 ± 28 vs. 532 ± 6, p < 0.05), significantly lower ACT compared to the baseline control (p < 0.05) and reduced pulmonary minute ventilation compared to pre-CNO control (p < 0.05). M3Dq DREADD activation did not cause bronchoconstriction (separate protocol), however, there was a concomitant reduction in HR, Tb and ventilation, accompanied by cardiac arrhythmias. We speculate that reductions in Tb, HR and ventilation reflect a mechanistic link between SKM Gq signaling and the metabolic responses associated with the initiation of torpor. Supported by the Canadian Institutes of Health Research (CIHR MOP-81211).
Collapse
Affiliation(s)
- Sandra G. Vincent
- Department of Biomedical and Molecular Sciences and Division of Respirology, Department of Medicine, Queen’s University, Kingston, ON K7L 3N6 Canada
| | - John T. Fisher
- Department of Biomedical and Molecular Sciences and Division of Respirology, Department of Medicine, Queen’s University, Kingston, ON K7L 3N6 Canada
| |
Collapse
|