1
|
Goldberg LR, Baskin BM, Beierle JA, Adla Y, Kelliher JC, Yao EJ, Kirkpatrick SL, Reed ER, Jenkins DF, Cox J, Luong AM, Luttik KP, Scotellaro JA, Drescher TA, Crotts SB, Yazdani N, Ferris MT, Johnson WE, Mulligan MK, Bryant CD. Atp1a2 and Kcnj9 Are Candidate Genes Underlying Sensitivity to Oxycodone-Induced Locomotor Activation and Withdrawal-Induced Anxiety-Like Behaviors in C57BL/6 Substrains. GENES, BRAIN, AND BEHAVIOR 2025; 24:e70009. [PMID: 39801366 PMCID: PMC11725984 DOI: 10.1111/gbb.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/22/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025]
Abstract
Opioid use disorder is heritable, yet its genetic etiology is largely unknown. C57BL/6J and C57BL/6NJ mouse substrains exhibit phenotypic diversity in the context of limited genetic diversity which together can facilitate genetic discovery. Here, we found C57BL/6NJ mice were less sensitive to oxycodone (OXY)-induced locomotor activation versus C57BL/6J mice in a conditioned place preference paradigm. Narrow-sense heritability of OXY-induced locomotor activity traits ranged from 0.22 to 0.31, implicating suitability for genetic analysis. Quantitative trait locus (QTL) mapping in an F2 cross identified a chromosome 1 QTL explaining 7%-12% of the variance in OXY locomotion and anxiety-like withdrawal in the elevated plus maze. A second QTL for EPM withdrawal behavior on chromosome 5 near Gabra2 (alpha-2 subunit of GABA-A receptor) explained 9% of the variance. To narrow the chromosome 1 locus, we generated recombinant lines spanning 163-181 Mb, captured the QTL for OXY locomotor traits and withdrawal, and fine-mapped a 2.45-Mb region (170.16-172.61 Mb). Transcriptome analysis identified five, localized striatal cis-eQTL transcripts and two were confirmed at the protein level (KCNJ9, ATP1A2). Kcnj9 codes for a potassium channel (GIRK3) that is a major effector of mu opioid receptor signaling. Atp1a2 codes for a subunit of a Na+/K+ ATPase enzyme that regulates neuronal excitability and shows functional adaptations following chronic opioid administration. To summarize, we identified two candidate genes underlying the physiological and behavioral properties of opioids, with direct preclinical relevance to investigators employing these widely used substrains and clinical relevance to human genetic studies of opioid use disorder.
Collapse
Affiliation(s)
- Lisa R. Goldberg
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug DiscoveryNortheastern UniversityBostonMassachusettsUSA
- Graduate Program in Biomolecular Pharmacology, Department of Pharmacology, Physiology & BiophysicsBoston University Chobanian and Avedisian School of MedicineBostonMassachusettsUSA
| | - Britahny M. Baskin
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug DiscoveryNortheastern UniversityBostonMassachusettsUSA
- T32 Training Program on Development of Medications for Substance Use Disorder, Center for Drug DiscoveryNortheastern UniversityBostonMassachusettsUSA
| | - Jacob A. Beierle
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug DiscoveryNortheastern UniversityBostonMassachusettsUSA
- Graduate Program in Biomolecular Pharmacology, Department of Pharmacology, Physiology & BiophysicsBoston University Chobanian and Avedisian School of MedicineBostonMassachusettsUSA
- Transformative Training Program in Addiction ScienceBoston UniversityBostonMassachusettsUSA
| | - Yahia Adla
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug DiscoveryNortheastern UniversityBostonMassachusettsUSA
| | - Julia C. Kelliher
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug DiscoveryNortheastern UniversityBostonMassachusettsUSA
| | - Emily J. Yao
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug DiscoveryNortheastern UniversityBostonMassachusettsUSA
| | - Stacey L. Kirkpatrick
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug DiscoveryNortheastern UniversityBostonMassachusettsUSA
| | - Eric R. Reed
- Graduate Program in BioinformaticsBoston UniversityBostonMassachusettsUSA
| | - David F. Jenkins
- Graduate Program in BioinformaticsBoston UniversityBostonMassachusettsUSA
| | - Jiayi Cox
- Genetics and Graduate Program in Genetics and Genomics, Program in Biomedical SciencesBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Alexander M. Luong
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug DiscoveryNortheastern UniversityBostonMassachusettsUSA
| | - Kimberly P. Luttik
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug DiscoveryNortheastern UniversityBostonMassachusettsUSA
| | - Julia A. Scotellaro
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug DiscoveryNortheastern UniversityBostonMassachusettsUSA
- Undergraduate Research Opportunity Program (UROP)Boston UniversityBostonMassachusettsUSA
| | - Timothy A. Drescher
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug DiscoveryNortheastern UniversityBostonMassachusettsUSA
| | - Sydney B. Crotts
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug DiscoveryNortheastern UniversityBostonMassachusettsUSA
| | - Neema Yazdani
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug DiscoveryNortheastern UniversityBostonMassachusettsUSA
- Graduate Program in Biomolecular Pharmacology, Department of Pharmacology, Physiology & BiophysicsBoston University Chobanian and Avedisian School of MedicineBostonMassachusettsUSA
- Transformative Training Program in Addiction ScienceBoston UniversityBostonMassachusettsUSA
| | - Martin T. Ferris
- Department of GeneticsUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - W. Evan Johnson
- Division of Infectious Disease, Department of Medicine, Center for Data ScienceRutgers UniversityNew BrunswickNew JerseyUSA
| | - Megan K. Mulligan
- Department of Genetics, Genomics, and InformaticsUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Camron D. Bryant
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug DiscoveryNortheastern UniversityBostonMassachusettsUSA
- T32 Training Program on Development of Medications for Substance Use Disorder, Center for Drug DiscoveryNortheastern UniversityBostonMassachusettsUSA
| |
Collapse
|
2
|
Kim HG, Berdasco C, Nairn AC, Kim Y. The WAVE complex in developmental and adulthood brain disorders. Exp Mol Med 2025:10.1038/s12276-024-01386-w. [PMID: 39774290 DOI: 10.1038/s12276-024-01386-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 10/09/2024] [Accepted: 10/31/2024] [Indexed: 01/11/2025] Open
Abstract
Actin polymerization and depolymerization are fundamental cellular processes required not only for the embryonic and postnatal development of the brain but also for the maintenance of neuronal plasticity and survival in the adult and aging brain. The orchestrated organization of actin filaments is controlled by various actin regulatory proteins. Wiskott‒Aldrich syndrome protein-family verprolin-homologous protein (WAVE) members are key activators of ARP2/3 complex-mediated actin polymerization. WAVE proteins exist as heteropentameric complexes together with regulatory proteins, including CYFIP, NCKAP, ABI and BRK1. The activity of the WAVE complex is tightly regulated by extracellular cues and intracellular signaling to execute its roles in specific intracellular events in brain cells. Notably, dysregulation of the WAVE complex and WAVE complex-mediated cellular processes confers vulnerability to a variety of brain disorders. De novo mutations in WAVE genes and other components of the WAVE complex have been identified in patients with developmental disorders such as intellectual disability, epileptic seizures, schizophrenia, and/or autism spectrum disorder. In addition, alterations in the WAVE complex are implicated in the pathophysiology of Alzheimer's disease and Parkinson's disease, as well as in behavioral adaptations to psychostimulants or maladaptive feeding.
Collapse
Affiliation(s)
- Hyung-Goo Kim
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, 08854, USA
| | - Clara Berdasco
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, 08854, USA
| | - Angus C Nairn
- Department of Psychiatry, Yale School of Medicine, Connecticut Mental Health Center, New Haven, CT, USA
| | - Yong Kim
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, 08854, USA.
- Brain Health Institute, Rutgers University, Piscataway, NJ, 08854, USA.
| |
Collapse
|
3
|
Wingfield KK, Misic T, Miracle SA, McDermott CS, Jain K, Abney NM, Richardson KT, Rubman MB, Beierle JA, Wachman EM, Bryant CD. The acoustic properties, syllable structure, and syllable sequences of ultrasonic vocalizations (USVs) during neonatal opioid withdrawal in FVB/N mouse substrains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.06.622304. [PMID: 39574631 PMCID: PMC11580921 DOI: 10.1101/2024.11.06.622304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Concomitant with the opioid epidemic, there has been a rise in pregnant women diagnosed with opioid use disorder and cases of infants born with neonatal opioid withdrawal syndrome (NOWS). NOWS refers to signs and symptoms following cessation of prenatal opioid exposure that comprise neurological, gastrointestinal, and autonomic system dysfunction. A critical indicator of NOWS severity is excessive, high-pitched crying. However, NOWS evaluation is, in large part, subjective, and additional cry features may not be easily recognized during clinical assessment. Thus, there is a need for more objective measures to determine NOWS severity. We used a third trimester-approximate opioid exposure paradigm to model NOWS traits in genetically similar inbred substrains of FVB/N mice (NJ, NCrl, NHsd, and NTac). Pups were injected twice daily from postnatal day 1 (P1) to P14 with morphine (10 mg/kg, s.c.) or saline (20 ml/g, s.c.). Because there were only very minor substrain differences in spontaneous withdrawal-induced ultrasonic vocalization (USV) profiles, we collapsed across substrains to evaluate the effects of morphine withdrawal on additional USV properties. We identified syllable sequences unique to morphine-withdrawn and saline-control FVB/N pups on P7 and P14. We also observed an effect of spontaneous morphine withdrawal on the acoustic properties of USVs and specific syllables on P7 and P14. Multiple withdrawal traits correlated with some acoustic properties of USVs and syllable type emission in morphine-withdrawn FVB/N pups on P7 and P14. These data provide an in-depth investigation of mouse USV syllable profiles and acoustic features during spontaneous neonatal opioid withdrawal in mice.
Collapse
Affiliation(s)
- Kelly K. Wingfield
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
- T32 Biomolecular Pharmacology Training Program, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
| | - Teodora Misic
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
| | - Sophia A. Miracle
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
- Graduate Program for Neuroscience, Boston University, Boston, MA USA
| | - Carly S. McDermott
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
| | - Kaahini Jain
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
| | - Nalia M. Abney
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
| | - Kayla T. Richardson
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
- Post-Baccalaureate Research Education Program, Boston University Chobanian & Avedisian School of Medicine
| | | | - Jacob A. Beierle
- T32 Biomolecular Pharmacology Training Program, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
- Transformative Training Program in Addiction Science, Boston University Chobanian & Avedisian School of Medicine
| | - Elisha M. Wachman
- Department of Pediatrics, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston MA USA
| | - Camron D. Bryant
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
| |
Collapse
|
4
|
Goldberg LR, Baskin BM, Adla Y, Beierle JA, Kelliher JC, Yao EJ, Kirkpatrick SL, Reed ER, Jenkins DF, Cox J, Luong AM, Luttik KP, Scotellaro JA, Drescher TA, Crotts SB, Yazdani N, Ferris MT, Johnson WE, Mulligan MK, Bryant CD. Atp1a2 and Kcnj9 are candidate genes underlying sensitivity to oxycodone-induced locomotor activation and withdrawal-induced anxiety-like behaviors in C57BL/6 substrains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.16.589731. [PMID: 38798314 PMCID: PMC11123399 DOI: 10.1101/2024.04.16.589731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Opioid use disorder is heritable, yet its genetic etiology is largely unknown. C57BL/6J and C57BL/6NJ mouse substrains exhibit phenotypic diversity in the context of limited genetic diversity which together can facilitate genetic discovery. Here, we found C57BL/6NJ mice were less sensitive to oxycodone (OXY)-induced locomotor activation versus C57BL/6J mice in a conditioned place preference paradigm. Narrow-sense heritability was estimated at 0.22-0.31, implicating suitability for genetic analysis. Quantitative trait locus (QTL) mapping in an F2 cross identified a chromosome 1 QTL explaining 7-12% of the variance in OXY locomotion and anxiety-like withdrawal in the elevated plus maze. A second QTL for EPM withdrawal behavior on chromosome 5 near Gabra2 (alpha-2 subunit of GABA-A receptor) explained 9% of the variance. To narrow the chromosome 1 locus, we generated recombinant lines spanning 163-181 Mb, captured the QTL for OXY locomotor traits and withdrawal, and fine-mapped a 2.45-Mb region (170.16-172.61 Mb). Transcriptome analysis identified five, localized striatal cis-eQTL transcripts and two were confirmed at the protein level (KCNJ9, ATP1A2). Kcnj9 codes for a potassium channel (GIRK3) that is a major effector of mu opioid receptor signaling. Atp1a2 codes for a subunit of a Na+/K+ ATPase enzyme that regulates neuronal excitability and shows functional adaptations following chronic opioid administration. To summarize, we identified two candidate genes underlying the physiological and behavioral properties of opioids, with direct preclinical relevance to investigators employing these widely used substrains and clinical relevance to human genetic studies of opioid use disorder.
Collapse
Affiliation(s)
- Lisa R. Goldberg
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
- Graduate Program in Biomolecular Pharmacology, Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA USA
| | - Britahny M. Baskin
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
- T32 Training Program on Development of Medications for Substance Use Disorder, Center for Drug Discovery, Northeastern University
| | - Yahia Adla
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
| | - Jacob A. Beierle
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
- Graduate Program in Biomolecular Pharmacology, Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA USA
- Transformative Training Program in Addiction Science, Boston University
| | - Julia C. Kelliher
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
| | - Emily J. Yao
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
| | - Stacey L. Kirkpatrick
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
| | - Eric R. Reed
- Graduate Program in Bioinformatics, Boston University, Boston, MA USA
| | - David F. Jenkins
- Graduate Program in Bioinformatics, Boston University, Boston, MA USA
| | - Jiayi Cox
- Genetics and Graduate Program in Genetics and Genomics, Program in Biomedical Sciences, Boston University Chobanian & Avedisian School of Medicine
| | - Alexander M. Luong
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
| | - Kimberly P. Luttik
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
| | - Julia A. Scotellaro
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
- Undergraduate Research Opportunity Program (UROP), Boston University
| | - Timothy A. Drescher
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
| | - Sydney B. Crotts
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
| | - Neema Yazdani
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
- Graduate Program in Biomolecular Pharmacology, Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA USA
- Transformative Training Program in Addiction Science, Boston University
| | - Martin T. Ferris
- Department of Genetics, University of North Carolina, Chapel Hill, NC USA
| | - W. Evan Johnson
- Division of Infectious Disease, Department of Medicine, Center for Data Science, Rutgers University, New Jersey, USA
| | - Megan K. Mulligan
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN USA
| | - Camron D. Bryant
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
- T32 Training Program on Development of Medications for Substance Use Disorder, Center for Drug Discovery, Northeastern University
| |
Collapse
|
5
|
Kim YJ, Kim K, Lee Y, Min HW, Ko YH, Lee BR, Hur KH, Kim SK, Lee SY, Jang CG. The mutated cytoplasmic fragile X messenger ribonucleoprotein 1 (FMR1)-interacting protein 2 (CYFIP2 S968F) regulates cocaine-induced reward behaviour and plasticity in the nucleus accumbens. Br J Pharmacol 2024; 181:3327-3345. [PMID: 38751203 DOI: 10.1111/bph.16427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/02/2024] [Accepted: 04/25/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND AND PURPOSE Cytoplasmic fragile X messenger ribonucleoprotein 1 (FMR1)-interacting protein 2 (CYFIP2), as a component of the Wiskott-Aldrich syndrome protein family verprolin-homologous protein (WAVE) regulatory complex, is involved in actin polymerization, contributing to neuronal development and structural plasticity. Mutating serine-968 to phenylalanine (S968F) in CYFIP2 causes an altered cocaine response in mice. The neuronal mechanisms underlying this response remain unknown. EXPERIMENTAL APPROACH We performed cocaine reward-related behavioural tests and examined changes in synaptic protein phenotypes and neuronal morphology in the nucleus accumbens (NAc), using CYFIP2 S968F knock-in mice to investigate the role of CYFIP2 in regulating cocaine reward. KEY RESULTS CYFIP2 S968F mutation attenuated cocaine-induced behavioural sensitization and conditioned place preference. Cocaine-induced c-Fos was not observed in the NAc of CYFIP2 S968F knock-in mice. However, c-Fos induction was still evident in the medial prefrontal cortex (mPFC). CYFIP2 S968F mutation altered cocaine-associated CYFIP2 signalling, glutamatergic protein expression and synaptic density in the NAc following cocaine exposure. To further determine the role of CYFIP2 in NAc neuronal activity and the mPFC projecting to the NAc activity-mediating reward response, we used optogenetic tools to stimulate the NAc or mPFC-NAc pathway and observed that optogenetic activation of the NAc or mPFC-NAc pathway induced reward-related behaviours. This effect was not observed in the S968F mutation in CYFIP2. CONCLUSION AND IMPLICATIONS These results suggest that CYFIP2 plays a role in controlling cocaine-mediated neuronal function and structural plasticity in the NAc, and that CYFIP2 could serve as a target for regulating cocaine reward.
Collapse
Affiliation(s)
- Young-Jung Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Kyungin Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Youyoung Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hee-Won Min
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yong-Hyun Ko
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Bo-Ram Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Kwang-Hyun Hur
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seon-Kyung Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seok-Yong Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
6
|
Deslauriers JC, Ghotkar RP, Russ LA, Jarman JA, Martin RM, Tippett RG, Sumathipala SH, Burton DF, Cole DC, Marsden KC. Cyfip2 controls the acoustic startle threshold through FMRP, actin polymerization, and GABA B receptor function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.22.573054. [PMID: 38187577 PMCID: PMC10769380 DOI: 10.1101/2023.12.22.573054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Animals process a constant stream of sensory input, and to survive they must detect and respond to dangerous stimuli while ignoring innocuous or irrelevant ones. Behavioral responses are elicited when certain properties of a stimulus such as its intensity or size reach a critical value, and such behavioral thresholds can be a simple and effective mechanism to filter sensory information. For example, the acoustic startle response is a conserved and stereotyped defensive behavior induced by sudden loud sounds, but dysregulation of the threshold to initiate this behavior can result in startle hypersensitivity that is associated with sensory processing disorders including schizophrenia and autism. Through a previous forward genetic screen for regulators of the startle threshold a nonsense mutation in Cytoplasmic Fragile X Messenger Ribonucleoprotein (FMRP)-interacting protein 2 (cyfip2) was found that causes startle hypersensitivity in zebrafish larvae, but the molecular mechanisms by which Cyfip2 establishes the acoustic startle threshold are unknown. Here we used conditional transgenic rescue and CRISPR/Cas9 to determine that Cyfip2 acts though both Rac1 and FMRP pathways, but not the closely related FXR1 or FXR2, to establish the acoustic startle threshold during early neurodevelopment. To identify proteins and pathways that may be downstream effectors of Rac1 and FMRP, we performed a candidate-based drug screen that indicated that Cyfip2 can also act acutely to maintain the startle threshold branched actin polymerization and N-methyl D-aspartate receptors (NMDARs). To complement this approach, we used unbiased discovery proteomics to determine that loss of Cyfip2 alters cytoskeletal and extracellular matrix components while also disrupting oxidative phosphorylation and GABA receptor signaling. Finally, we functionally validated our proteomics findings by showing that activating GABAB receptors, which like NMDARs are also FMRP targets, restores normal startle sensitivity in cyfip2 mutants. Together, these data reveal multiple mechanisms by which Cyfip2 regulates excitatory/inhibitory balance in the startle circuit to control the processing of acoustic information.
Collapse
Affiliation(s)
- Jacob C. Deslauriers
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Rohit P. Ghotkar
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Current address: Putnam Associates, Boston, Massachusetts, USA
| | - Lindsey A. Russ
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Current address: Department of Pharmacology & Physiology, Georgetown University, Washington D.C., USA
| | - Jordan A. Jarman
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Current address: Department of Physiology and Biophysics, Boston University, Boston, MA, USA
| | - Rubia M. Martin
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Current address: U.S. Environmental Protection Agency, Raleigh-Durham-Chapel Hill, North Carolina, USA
| | - Rachel G. Tippett
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Sureni H. Sumathipala
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Derek F. Burton
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - D. Chris Cole
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Kurt C. Marsden
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Center for Human Health and the Environment (CHHE), North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
7
|
Bagley JR, Tan Y, Zhu W, Cheng Z, Takeda S, Fang Z, Arslan A, Wang M, Guan Y, Jiang L, Jian R, Gu F, Parada I, Prince D, Jentsch JD, Peltz G. Neuron Navigator 1 (Nav1) regulates the response to cocaine in mice. Commun Biol 2023; 6:1053. [PMID: 37853211 PMCID: PMC10584906 DOI: 10.1038/s42003-023-05430-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/06/2023] [Indexed: 10/20/2023] Open
Abstract
Genetic variation accounts for much of the risk for developing a substance use disorder, but the underlying genetic factors and their genetic effector mechanisms are mostly unknown. Inbred mouse strains exhibit substantial and heritable differences in the extent of voluntary cocaine self-administration. Computational genetic analysis of cocaine self-administration data obtained from twenty-one inbred strains identified Nav1, a member of the neuron navigator family that regulates dendrite formation and axonal guidance, as a candidate gene. To test this genetic hypothesis, we generated and characterized Nav1 knockout mice. Consistent with the genetic prediction, Nav1 knockout mice exhibited increased voluntary cocaine intake and had increased motivation for cocaine consumption. Immunohistochemistry, electrophysiology, and transcriptomic studies were performed as a starting point for investigating the mechanism for the Nav1 knockout effect. Nav1 knockout mice had a reduced inhibitory synapse density in their cortex, increased excitatory synaptic transmission in their cortex and hippocampus, and increased excitatory neurons in a deep cortical layer. Collectively, our results indicate that Nav1 regulates the response to cocaine, and we identified Nav1 knockout induced changes in the excitatory and inhibitory synaptic balance in the cortex and hippocampus that could contribute to this effect.
Collapse
Affiliation(s)
- Jared R Bagley
- Department of Psychology, Binghamton University, Binghamton, NY, USA
| | - Yalun Tan
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Wan Zhu
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Zhuanfen Cheng
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Saori Takeda
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Zhouqing Fang
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Ahmed Arslan
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Meiyue Wang
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Yuan Guan
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Lihua Jiang
- Department of Genetics, Stanford University Medical School, Stanford, CA, USA
| | - Ruiqi Jian
- Department of Genetics, Stanford University Medical School, Stanford, CA, USA
| | - Feng Gu
- Department of Neurology, Stanford University Medical School, Stanford, CA, USA
- Department of Biological Sciences, University of North Texas, Denton, USA
| | - Isabel Parada
- Department of Neurology, Stanford University Medical School, Stanford, CA, USA
| | - David Prince
- Department of Neurology, Stanford University Medical School, Stanford, CA, USA
| | - J David Jentsch
- Department of Psychology, Binghamton University, Binghamton, NY, USA
| | - Gary Peltz
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA.
| |
Collapse
|
8
|
Kramer DA, Narvaez-Ortiz HY, Patel U, Shi R, Shen K, Nolen BJ, Roche J, Chen B. The intrinsically disordered cytoplasmic tail of a dendrite branching receptor uses two distinct mechanisms to regulate the actin cytoskeleton. eLife 2023; 12:e88492. [PMID: 37555826 PMCID: PMC10411975 DOI: 10.7554/elife.88492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/01/2023] [Indexed: 08/10/2023] Open
Abstract
Dendrite morphogenesis is essential for neural circuit formation, yet the molecular mechanisms underlying complex dendrite branching remain elusive. Previous studies on the highly branched Caenorhabditis elegans PVD sensory neuron identified a membrane co-receptor complex that links extracellular signals to intracellular actin remodeling machinery, promoting high-order dendrite branching. In this complex, the claudin-like transmembrane protein HPO-30 recruits the WAVE regulatory complex (WRC) to dendrite branching sites, stimulating the Arp2/3 complex to polymerize actin. We report here our biochemical and structural analysis of this interaction, revealing that the intracellular domain (ICD) of HPO-30 is intrinsically disordered and employs two distinct mechanisms to regulate the actin cytoskeleton. First, HPO-30 ICD binding to the WRC requires dimerization and involves the entire ICD sequence, rather than a short linear peptide motif. This interaction enhances WRC activation by the GTPase Rac1. Second, HPO-30 ICD directly binds to the sides and barbed end of actin filaments. Binding to the barbed end requires ICD dimerization and inhibits both actin polymerization and depolymerization, resembling the actin capping protein CapZ. These dual functions provide an intriguing model of how membrane proteins can integrate distinct mechanisms to fine-tune local actin dynamics.
Collapse
Affiliation(s)
- Daniel A Kramer
- Roy J Carver Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State UniversityAmesUnited States
| | - Heidy Y Narvaez-Ortiz
- Department of Chemistry and Biochemistry, Institute of Molecular Biology, University of OregonEugeneUnited States
| | - Urval Patel
- Roy J Carver Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State UniversityAmesUnited States
| | - Rebecca Shi
- Department of Biology, Stanford UniversityStanfordUnited States
- Neurosciences IDP, Stanford UniversityStanfordUnited States
| | - Kang Shen
- Department of Biology, Stanford UniversityStanfordUnited States
- Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Brad J Nolen
- Department of Chemistry and Biochemistry, Institute of Molecular Biology, University of OregonEugeneUnited States
| | - Julien Roche
- Roy J Carver Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State UniversityAmesUnited States
| | - Baoyu Chen
- Roy J Carver Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State UniversityAmesUnited States
| |
Collapse
|
9
|
Hartmann MC, McCulley WD, Holbrook SE, Haney MM, Smith CG, Kumar V, Rosenwasser AM. Cyfip2 allelic variation in C57BL/6J and C57BL/6NJ mice alters free-choice ethanol drinking but not binge-like drinking or wheel-running activity. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:1518-1529. [PMID: 37356964 DOI: 10.1111/acer.15137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 06/17/2023] [Accepted: 06/21/2023] [Indexed: 06/27/2023]
Abstract
BACKGROUND Since the origin of the C57BL/6 (B6) mouse strain, several phenotypically and genetically distinct B6 substrains have emerged. For example, C57BL/6J mice (B6J) display greater voluntary ethanol consumption and locomotor response to psychostimulants and differences in nucleus accumbens synaptic physiology relative to C57BL/6N (B6N) mice. A non-synonymous serine to phenylalanine point mutation (S968F) in the cytoplasmic FMR1-interacting protein 2 (Cyfip2) gene underlies both the differential locomotor response to cocaine and the accumbal physiology exhibited by these substrains. We examined whether Cyfip2 allelic variation underlies B6 substrain differences in other reward-related phenotypes, such as ethanol intake and wheel-running activity. METHODS We compared voluntary ethanol consumption, wheel-running, and binge-like ethanol drinking in male and female B6J and B6NJ mice. When substrain differences were observed, additional experiments were performed in two novel mouse models in which the B6N Cyfip2 mutation was either introduced (S968F) into the B6J background or corrected (F968S) via CRISPR/Cas9 technology. RESULTS B6J consumed significantly more ethanol than B6NJ and allelic variation in Cyfip2 contributed substantially to this substrain difference. In contrast, B6NJ displayed significantly more daily wheel-running than B6J, with Cyfip2 allelic variation playing only a minor role in this substrain difference. Lastly, no substrain differences were observed in binge-like ethanol drinking. CONCLUSIONS These results contribute to the characterization of behavior-genetic differences between B6 substrains, support previous work indicating that free-choice and binge-like ethanol drinking are dependent on partially distinct genetic networks, and identify a novel phenotypic difference between B6 substrains in wheel-running activity.
Collapse
Affiliation(s)
- Matthew C Hartmann
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, Maine, USA
- Department of Psychology, University of Maine, Orono, Maine, USA
| | | | - Sarah E Holbrook
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, Maine, USA
- The Jackson Laboratory, Bar Harbor, Maine, USA
| | - Megan M Haney
- Department of Psychology, University of Maine, Orono, Maine, USA
| | - Caitlin G Smith
- Department of Psychology, University of Maine, Orono, Maine, USA
| | - Vivek Kumar
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, Maine, USA
- The Jackson Laboratory, Bar Harbor, Maine, USA
| | - Alan M Rosenwasser
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, Maine, USA
- Department of Psychology, University of Maine, Orono, Maine, USA
- School of Biology and Ecology, University of Maine, Orono, Maine, USA
| |
Collapse
|
10
|
Ratković D, Knežević V, Dickov A, Fedrigolli E, Čomić M. Comparison of binge-eating disorder and food addiction. J Int Med Res 2023; 51:3000605231171016. [PMID: 37115520 PMCID: PMC10155018 DOI: 10.1177/03000605231171016] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023] Open
Abstract
In the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition, binge-eating disorder (BED) is classified as episodes of binge eating while not being hungry, eating too fast until feeling uncomfortably full, or eating in solitude with feelings of shame and disgust after eating, without compensatory mechanisms. The controversial disorder food addiction (FA) is characterized by overconsumption, cravings, failure to cut down on amounts of food, and withdrawal and tolerance to overeating. In this narrative review, we aimed to comprehensively characterize and compare BED and FA. We searched PubMed using the keywords "binge-eating disorder" and "food addiction." We finally included 51 publications according to topic specificity, credibility, the authors' reputation, and non-bias criteria. BED is characterized by concerns about dietary issues, body shape, and weight as well as depressive symptoms and brooding rumination. FA can be divided into substance addiction and behavioral addiction, which can be differentiated using a list of criteria including hunger, taste, pleasure, function of food, loss of social connections, weight concerns, and awareness about the disorder. Further research is needed to further characterize and distinguish BED and FA.
Collapse
Affiliation(s)
- Dragana Ratković
- University of Novi Sad, Faculty of Medicine Novi Sad, Department of Psychiatry and Psychological Medicine, Novi Sad, Republic of Serbia
- Psychiatry Clinic, University Clinical Center of Vojvodina, Novi Sad, Republic of Serbia
| | - Vladimir Knežević
- University of Novi Sad, Faculty of Medicine Novi Sad, Department of Psychiatry and Psychological Medicine, Novi Sad, Republic of Serbia
- Psychiatry Clinic, University Clinical Center of Vojvodina, Novi Sad, Republic of Serbia
| | - Aleksandra Dickov
- University of Novi Sad, Faculty of Medicine Novi Sad, Department of Psychiatry and Psychological Medicine, Novi Sad, Republic of Serbia
- Psychiatry Clinic, University Clinical Center of Vojvodina, Novi Sad, Republic of Serbia
| | - Elsa Fedrigolli
- University of Novi Sad, Faculty of Medicine Novi Sad, Department of Psychiatry and Psychological Medicine, Novi Sad, Republic of Serbia
| | - Maša Čomić
- University of Novi Sad, Faculty of Medicine Novi Sad, Department of Psychiatry and Psychological Medicine, Novi Sad, Republic of Serbia
- Psychiatry Clinic, University Clinical Center of Vojvodina, Novi Sad, Republic of Serbia
| |
Collapse
|
11
|
Hellbach F, Sinke L, Costeira R, Baumeister SE, Beekman M, Louca P, Leeming ER, Mompeo O, Berry S, Wilson R, Wawro N, Freuer D, Hauner H, Peters A, Winkelmann J, Koenig W, Meisinger C, Waldenberger M, Heijmans BT, Slagboom PE, Bell JT, Linseisen J. Pooled analysis of epigenome-wide association studies of food consumption in KORA, TwinsUK and LLS. Eur J Nutr 2023; 62:1357-1375. [PMID: 36571600 PMCID: PMC10030421 DOI: 10.1007/s00394-022-03074-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/12/2022] [Indexed: 12/27/2022]
Abstract
PURPOSE Examining epigenetic patterns is a crucial step in identifying molecular changes of disease pathophysiology, with DNA methylation as the most accessible epigenetic measure. Diet is suggested to affect metabolism and health via epigenetic modifications. Thus, our aim was to explore the association between food consumption and DNA methylation. METHODS Epigenome-wide association studies were conducted in three cohorts: KORA FF4, TwinsUK, and Leiden Longevity Study, and 37 dietary exposures were evaluated. Food group definition was harmonized across the three cohorts. DNA methylation was measured using Infinium MethylationEPIC BeadChip in KORA and Infinium HumanMethylation450 BeadChip in the Leiden study and the TwinsUK study. Overall, data from 2293 middle-aged men and women were included. A fixed-effects meta-analysis pooled study-specific estimates. The significance threshold was set at 0.05 for false-discovery rate-adjusted p values per food group. RESULTS We identified significant associations between the methylation level of CpG sites and the consumption of onions and garlic (2), nuts and seeds (18), milk (1), cream (11), plant oils (4), butter (13), and alcoholic beverages (27). The signals targeted genes of metabolic health relevance, for example, GLI1, RPTOR, and DIO1, among others. CONCLUSION This EWAS is unique with its focus on food groups that are part of a Western diet. Significant findings were mostly related to food groups with a high-fat content.
Collapse
Affiliation(s)
- Fabian Hellbach
- Institute for Medical Information Processing, Biometry, and Epidemiology, Medical Faculty, Ludwig-Maximilian University Munich, Marchioninistr. 15, 81377, Munich, Germany.
- Epidemiology, Faculty of Medicine, University of Augsburg, University Hospital Augsburg, Stenglinstraße 2, 86156, Augsburg, Germany.
| | - Lucy Sinke
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
| | - Ricardo Costeira
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, England, UK
| | - Sebastian-Edgar Baumeister
- Institute of Health Services Research in Dentistry, Medical Faculty, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Marian Beekman
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
| | - Panayiotis Louca
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, England, UK
| | - Emily R Leeming
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, England, UK
| | - Olatz Mompeo
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, England, UK
| | - Sarah Berry
- Department of Nutritional Sciences, King's College London, London, UK
| | - Rory Wilson
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Nina Wawro
- Institute for Medical Information Processing, Biometry, and Epidemiology, Medical Faculty, Ludwig-Maximilian University Munich, Marchioninistr. 15, 81377, Munich, Germany
- Epidemiology, Faculty of Medicine, University of Augsburg, University Hospital Augsburg, Stenglinstraße 2, 86156, Augsburg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Dennis Freuer
- Epidemiology, Faculty of Medicine, University of Augsburg, University Hospital Augsburg, Stenglinstraße 2, 86156, Augsburg, Germany
| | - Hans Hauner
- Else Kröner-Fresenius-Center for Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, 85354, Freising, Germany
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, Georg-Brauchle-Ring 62, 80992, Munich, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD E.V.), Ingolstädter Landstr. 1, 85764, Munich-Neuherberg, Germany
| | - Juliane Winkelmann
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Center for Environmental Health (HmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Wolfgang Koenig
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Pettenkoferstr. 8A & 9, 80336, Munich, Germany
- German Heart Centre Munich, Technical University Munich, Lazarettstr. 36, 80636, Munich, Germany
- Institute of Epidemiology and Medical Biometry, University of Ulm, Helmholtzstr. 22, 89081, Ulm, Germany
| | - Christa Meisinger
- Epidemiology, Faculty of Medicine, University of Augsburg, University Hospital Augsburg, Stenglinstraße 2, 86156, Augsburg, Germany
| | - Melanie Waldenberger
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD E.V.), Ingolstädter Landstr. 1, 85764, Munich-Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Pettenkoferstr. 8A & 9, 80336, Munich, Germany
| | - Bastiaan T Heijmans
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
| | - P Eline Slagboom
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
| | - Jordana T Bell
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, England, UK
| | - Jakob Linseisen
- Institute for Medical Information Processing, Biometry, and Epidemiology, Medical Faculty, Ludwig-Maximilian University Munich, Marchioninistr. 15, 81377, Munich, Germany
- Epidemiology, Faculty of Medicine, University of Augsburg, University Hospital Augsburg, Stenglinstraße 2, 86156, Augsburg, Germany
| |
Collapse
|
12
|
Ousey J, Boktor JC, Mazmanian SK. Gut microbiota suppress feeding induced by palatable foods. Curr Biol 2023; 33:147-157.e7. [PMID: 36450285 PMCID: PMC9839363 DOI: 10.1016/j.cub.2022.10.066] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 08/30/2022] [Accepted: 10/28/2022] [Indexed: 12/03/2022]
Abstract
Feeding behaviors depend on intrinsic and extrinsic factors including genetics, food palatability, and the environment.1,2,3,4,5 The gut microbiota is a major environmental contributor to host physiology and impacts feeding behavior.6,7,8,9,10,11,12 Here, we explored the hypothesis that gut bacteria influence behavioral responses to palatable foods and reveal that antibiotic depletion (ABX) of the gut microbiota in mice results in overconsumption of several palatable foods with conserved effects on feeding dynamics. Gut microbiota restoration via fecal transplant into ABX mice is sufficient to rescue overconsumption of high-sucrose pellets. Operant conditioning tests found that ABX mice exhibit intensified motivation to pursue high-sucrose rewards. Accordingly, neuronal activity in mesolimbic brain regions, which have been linked with motivation and reward-seeking behavior,3 was elevated in ABX mice after consumption of high-sucrose pellets. Differential antibiotic treatment and functional microbiota transplants identified specific gut bacterial taxa from the family S24-7 and the genus Lactobacillus whose abundances associate with suppression of high-sucrose pellet consumption. Indeed, colonization of mice with S24-7 and Lactobacillus johnsonii was sufficient to reduce overconsumption of high-sucrose pellets in an antibiotic-induced model of binge eating. These results demonstrate that extrinsic influences from the gut microbiota can suppress the behavioral response toward palatable foods in mice.
Collapse
Affiliation(s)
- James Ousey
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E California Blvd, Pasadena, CA 91125, USA.
| | - Joseph C Boktor
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E California Blvd, Pasadena, CA 91125, USA
| | - Sarkis K Mazmanian
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E California Blvd, Pasadena, CA 91125, USA.
| |
Collapse
|
13
|
Brain serotonin deficiency and fluoxetine lead to sex-specific effects on binge-like food consumption in mice. Psychopharmacology (Berl) 2022; 239:2975-2984. [PMID: 35750862 DOI: 10.1007/s00213-022-06181-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/17/2022] [Indexed: 10/17/2022]
Abstract
RATIONALE Although pharmacotherapies are often effective in reducing binge eating in conditions such as bulimia nervosa and binge eating disorder, subsets of patients do not benefit sufficiently from existing treatments, and the reasons for treatment failure remain unclear. OBJECTIVES This study aimed to evaluate whether genetic reductions in brain serotonin influence binge eating and/or the ability of fluoxetine, a selective serotonin reuptake inhibitor, to reduce binge eating in mice. METHODS This study used a validated model of binge-like consumption of high-fat diet to compare binge-like food intake in control and fluoxetine-treated wild-type and serotonin-deficient mice from the tryptophan hydroxylase 2 (R439H) knock-in line. In addition, real-time PCR was used to evaluate potential genotype and sex differences in the effects of fluoxetine on gene expression in the raphe nucleus. RESULTS The results reveal that brain serotonin deficiency is sufficient to increase binge eating in males, but not females. However, while chronic fluoxetine reduced binge eating in both genotypes of males and in wild-type females, it failed to reduce binge eating in serotonin-deficient females. Transcriptional responses to chronic fluoxetine were also characterized by sex and genotype differences. CONCLUSIONS Overall, this study revealed significant sex differences in the effects of fluoxetine and brain serotonin deficiency on binge-like food intake and suggests that low brain serotonin could impact eating disorders both by promoting binge eating and by limiting the efficacy of fluoxetine to reduce binge eating.
Collapse
|
14
|
Beierle JA, Yao EJ, Goldstein SI, Lynch WB, Scotellaro JL, Shah AA, Sena KD, Wong AL, Linnertz CL, Averin O, Moody DE, Reilly CA, Peltz G, Emili A, Ferris MT, Bryant CD. Zhx2 Is a Candidate Gene Underlying Oxymorphone Metabolite Brain Concentration Associated with State-Dependent Oxycodone Reward. J Pharmacol Exp Ther 2022; 382:167-180. [PMID: 35688478 PMCID: PMC9341249 DOI: 10.1124/jpet.122.001217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/16/2022] [Indexed: 11/22/2022] Open
Abstract
Understanding the pharmacogenomics of opioid metabolism and behavior is vital to therapeutic success, as mutations can dramatically alter therapeutic efficacy and addiction liability. We found robust, sex-dependent BALB/c substrain differences in oxycodone behaviors and whole brain concentration of oxycodone metabolites. BALB/cJ females showed robust state-dependent oxycodone reward learning as measured via conditioned place preference when compared with the closely related BALB/cByJ substrain. Accordingly, BALB/cJ females also showed a robust increase in brain concentration of the inactive metabolite noroxycodone and the active metabolite oxymorphone compared with BALB/cByJ mice. Oxymorphone is a highly potent, full agonist at the mu opioid receptor that could enhance drug-induced interoception and state-dependent oxycodone reward learning. Quantitative trait locus (QTL) mapping in a BALB/c F2 reduced complexity cross revealed one major QTL on chromosome 15 underlying brain oxymorphone concentration that explained 32% of the female variance. BALB/cJ and BALB/cByJ differ by fewer than 10,000 variants, which can greatly facilitate candidate gene/variant identification. Hippocampal and striatal cis-expression QTL (eQTL) and exon-level eQTL analysis identified Zhx2, a candidate gene coding for a transcriptional repressor with a private BALB/cJ retroviral insertion that reduces Zhx2 expression and sex-dependent dysregulation of cytochrome P450 enzymes. Whole brain proteomics corroborated the Zhx2 eQTL and identified upregulated CYP2D11 that could increase brain oxymorphone in BALB/cJ females. To summarize, Zhx2 is a highly promising candidate gene underlying brain oxycodone metabolite levels. Future studies will validate Zhx2 and its site of action using reciprocal gene editing and tissue-specific viral manipulations in BALB/c substrains. SIGNIFICANCE STATEMENT: Our findings show that genetic variation can result in sex-specific alterations in whole brain concentration of a bioactive opioid metabolite after oxycodone administration, reinforcing the need for sex as a biological factor in pharmacogenomic studies. The cooccurrence of female-specific increased oxymorphone and state-dependent reward learning suggests that this minor yet potent and efficacious metabolite of oxycodone could increase opioid interoception and drug-cue associative learning of opioid reward, which has implications for cue-induced relapse of drug-seeking behavior and for precision pharmacogenetics.
Collapse
Affiliation(s)
- Jacob A Beierle
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Emily J Yao
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Stanley I Goldstein
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - William B Lynch
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Julia L Scotellaro
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Anyaa A Shah
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Katherine D Sena
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Alyssa L Wong
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Colton L Linnertz
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Olga Averin
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - David E Moody
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Christopher A Reilly
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Gary Peltz
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Andrew Emili
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Martin T Ferris
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Camron D Bryant
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| |
Collapse
|
15
|
Sena KD, Beierle JA, Richardson KT, Kantak KM, Bryant CD. Assessment of Binge-Like Eating of Unsweetened vs. Sweetened Chow Pellets in BALB/c Substrains. Front Behav Neurosci 2022; 16:944890. [PMID: 35910681 PMCID: PMC9337213 DOI: 10.3389/fnbeh.2022.944890] [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: 05/16/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Binge eating disorder (BED) is defined as chronic episodes of consuming large amounts of food in less than 2 h. Binge eating disorder poses a serious public health problem, as it increases the risk of obesity, type II diabetes, and heart disease. Binge eating is a highly heritable trait; however, its genetic basis remains largely unexplored. We employed a mouse model for binge eating that focused on identifying heritable differences between inbred substrains in acute and escalated intake of sucrose-sweetened palatable food vs. unsweetened chow pellets in a limited, intermittent access paradigm. In the present study, we examined two genetically similar substrains of BALB/c mice for escalation in food consumption, incubation of craving after a no-food training period, and compulsive-like food consumption in an aversive context. BALB/cJ and BALB/cByJ mice showed comparable levels of acute and escalated consumption of palatable food across training trials. Surprisingly, BALB/cByJ mice also showed binge-like eating of the unsweetened chow pellets similar to the escalation in palatable food intake of both substrains. Finally, we replicated the well-documented decrease in anxiety-like behavior in BALB/cByJ mice in the light-dark conflict test that likely contributed to greater palatable food intake than BALB/cJ in the light arena. To summarize, BALB/cByJ mice show binge-like eating in the presence and absence of sucrose. Possible explanations for the lack of selectivity in binge-like eating across diets (e.g., novelty preference, taste) are discussed.
Collapse
Affiliation(s)
- Katherine D. Sena
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Jacob A. Beierle
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Kayla T. Richardson
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Kathleen M. Kantak
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, United States
| | - Camron D. Bryant
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
16
|
Akinola LS, Rahman Y, Ondo O, Gonzales J, Bagdas D, Jackson A, Davidson-Wert N, Damaj MI. Genotypic Differences in the Effects of Menthol on Nicotine Intake and Preference in Mice. Front Neurosci 2022; 16:905330. [PMID: 35769694 PMCID: PMC9234577 DOI: 10.3389/fnins.2022.905330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
Menthol has been shown to exacerbate elements of nicotine addiction in humans and rodents; however, the mechanisms mediating its effects are not fully understood. This study examined the impact of genetic factors in menthol’s effects on oral nicotine consumption by comparing two inbred mouse strains with differing sensitivities to nicotine. C57BL/6J (B6J) mice are nicotine-preferring, while DBA/2J (D2J) mice are not. While the effects of menthol on oral nicotine consumption have been highlighted in B6J mice, it is unknown if they extend to the D2J strain as well. Consequently, adolescent (PND 21) and adult (PND 63), male and female D2J mice were subjected to the nicotine two-bottle choice (2BC) paradigm with orally and systemically administered menthol. Then, we evaluated its impact on nicotine pharmacological responses in conditioned reward and nociception after systemic administration and, lastly, investigated the potential involvement of the TAAR1 gene and α7 nAChRs in menthol’s effects. Menthol failed to enhance oral nicotine consumption in adult and adolescent female and male D2J mice. Moreover, this lack in effect was not due to nicotine concentration, oral aversion to menthol, or basal preference for nicotine. Menthol also failed to augment nicotine reward or enhance nicotine-induced antinociception in D2J mice, demonstrating that genetic background plays a significant role in sensitivity to menthol’s effects on nicotine. Furthermore, TAAR1 or α7 nAChRs did not seem to mediate menthol’s differential effects in D2J mice. These findings support the existence of genotype-specific mechanisms that may contribute to the variable effects of menthol in different populations.
Collapse
Affiliation(s)
- Lois S. Akinola
- Department of Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, United States
- *Correspondence: Lois S. Akinola,
| | - Yumna Rahman
- Department of Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, United States
| | - Olivia Ondo
- Department of Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, United States
| | - Jada Gonzales
- Department of Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, United States
| | - Deniz Bagdas
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, United States
- Yale Tobacco Center of Regulatory Science, Yale School of Medicine, New Haven, CT, United States
| | - Asti Jackson
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, United States
- Yale Tobacco Center of Regulatory Science, Yale School of Medicine, New Haven, CT, United States
| | - Nicole Davidson-Wert
- Department of Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, United States
| | - M. Imad Damaj
- Department of Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
17
|
Kramer DA, Piper HK, Chen B. WASP family proteins: Molecular mechanisms and implications in human disease. Eur J Cell Biol 2022; 101:151244. [PMID: 35667337 PMCID: PMC9357188 DOI: 10.1016/j.ejcb.2022.151244] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 02/08/2023] Open
Abstract
Proteins of the Wiskott-Aldrich syndrome protein (WASP) family play a central role in regulating actin cytoskeletal dynamics in a wide range of cellular processes. Genetic mutations or misregulation of these proteins are tightly associated with many diseases. The WASP-family proteins act by transmitting various upstream signals to their conserved WH2-Central-Acidic (WCA) peptide sequence at the C-terminus, which in turn binds to the Arp2/3 complex to stimulate the formation of branched actin networks at membranes. Despite this common feature, the regulatory mechanisms and cellular functions of distinct WASP-family proteins are very different. Here, we summarize and clarify our current understanding of WASP-family proteins and how disruption of their functions is related to human disease.
Collapse
Affiliation(s)
- Daniel A Kramer
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, 2437 Pammel Drive, Ames, IA 50011, USA
| | - Hannah K Piper
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, 2437 Pammel Drive, Ames, IA 50011, USA
| | - Baoyu Chen
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, 2437 Pammel Drive, Ames, IA 50011, USA.
| |
Collapse
|
18
|
Gaines CH, Schoenrock SA, Farrington J, Lee DF, Aponte-Collazo LJ, Shaw GD, Miller DR, Ferris MT, Pardo-Manuel de Villena F, Tarantino LM. Cocaine-Induced Locomotor Activation Differs Across Inbred Mouse Substrains. Front Psychiatry 2022; 13:800245. [PMID: 35599758 PMCID: PMC9120424 DOI: 10.3389/fpsyt.2022.800245] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
Cocaine use disorders (CUD) are devastating for affected individuals and impose a significant societal burden, but there are currently no FDA-approved therapies. The development of novel and effective treatments has been hindered by substantial gaps in our knowledge about the etiology of these disorders. The risk for developing a CUD is influenced by genetics, the environment and complex interactions between the two. Identifying specific genes and environmental risk factors that increase CUD risk would provide an avenue for the development of novel treatments. Rodent models of addiction-relevant behaviors have been a valuable tool for studying the genetics of behavioral responses to drugs of abuse. Traditional genetic mapping using genetically and phenotypically divergent inbred mice has been successful in identifying numerous chromosomal regions that influence addiction-relevant behaviors, but these strategies rarely result in identification of the causal gene or genetic variant. To overcome this challenge, reduced complexity crosses (RCC) between closely related inbred mouse strains have been proposed as a method for rapidly identifying and validating functional variants. The RCC approach is dependent on identifying phenotypic differences between substrains. To date, however, the study of addiction-relevant behaviors has been limited to very few sets of substrains, mostly comprising the C57BL/6 lineage. The present study expands upon the current literature to assess cocaine-induced locomotor activation in 20 inbred mouse substrains representing six inbred strain lineages (A/J, BALB/c, FVB/N, C3H/He, DBA/2 and NOD) that were either bred in-house or supplied directly by a commercial vendor. To our knowledge, we are the first to identify significant differences in cocaine-induced locomotor response in several of these inbred substrains. The identification of substrain differences allows for the initiation of RCC populations to more rapidly identify specific genetic variants associated with acute cocaine response. The observation of behavioral profiles that differ between mice generated in-house and those that are vendor-supplied also presents an opportunity to investigate the influence of environmental factors on cocaine-induced locomotor activity.
Collapse
Affiliation(s)
- Christiann H. Gaines
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Neuroscience Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Sarah A. Schoenrock
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Joseph Farrington
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - David F. Lee
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Pharmacology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lucas J. Aponte-Collazo
- Pharmacology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ginger D. Shaw
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Darla R. Miller
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Martin T. Ferris
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Fernando Pardo-Manuel de Villena
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lisa M. Tarantino
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
19
|
Bulik CM, Coleman JRI, Hardaway JA, Breithaupt L, Watson HJ, Bryant CD, Breen G. Genetics and neurobiology of eating disorders. Nat Neurosci 2022; 25:543-554. [PMID: 35524137 PMCID: PMC9744360 DOI: 10.1038/s41593-022-01071-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 04/01/2022] [Indexed: 12/14/2022]
Abstract
Eating disorders (anorexia nervosa, bulimia nervosa and binge-eating disorder) are a heterogeneous class of complex illnesses marked by weight and appetite dysregulation coupled with distinctive behavioral and psychological features. Our understanding of their genetics and neurobiology is evolving thanks to global cooperation on genome-wide association studies, neuroimaging, and animal models. Until now, however, these approaches have advanced the field in parallel, with inadequate cross-talk. This review covers overlapping advances in these key domains and encourages greater integration of hypotheses and findings to create a more unified science of eating disorders. We highlight ongoing and future work designed to identify implicated biological pathways that will inform staging models based on biology as well as targeted prevention and tailored intervention, and will galvanize interest in the development of pharmacologic agents that target the core biology of the illnesses, for which we currently have few effective pharmacotherapeutics.
Collapse
Affiliation(s)
- Cynthia M Bulik
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| | - Jonathan R I Coleman
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- National Institute of Health Research Maudsley Biomedical Research Centre, South London and Maudsley National Health Service Trust, London, UK
| | - J Andrew Hardaway
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lauren Breithaupt
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Eating Disorders Clinical and Research Program, Massachusetts General Hospital, Boston, MA, USA
| | - Hunna J Watson
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- School of Psychology, Curtin University, Perth, Western Australia, Australia
- Division of Paediatrics, School of Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Camron D Bryant
- Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Gerome Breen
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- National Institute of Health Research Maudsley Biomedical Research Centre, South London and Maudsley National Health Service Trust, London, UK
| |
Collapse
|
20
|
Honeywell KM, Doren EV, Szumlinski KK. Selective Inhibition of PDE4B Reduces Methamphetamine Reinforcement in Two C57BL/6 Substrains. Int J Mol Sci 2022; 23:4872. [PMID: 35563262 PMCID: PMC9099926 DOI: 10.3390/ijms23094872] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/13/2022] [Accepted: 04/26/2022] [Indexed: 12/02/2022] Open
Abstract
Methamphetamine (MA) is a highly addictive psychostimulant drug, and the number of MA-related overdose deaths has reached epidemic proportions. Repeated MA exposure induces a robust and persistent neuroinflammatory response, and the evidence supports the potential utility of targeting neuroimmune function using non-selective phosphodiesterase 4 (PDE4) inhibitors as a therapeutic strategy for attenuating addiction-related behavior. Off-target, emetic effects associated with non-selective PDE4 blockade led to the development of isozyme-selective inhibitors, of which the PDE4B-selective inhibitor A33 was demonstrated recently to reduce binge drinking in two genetically related C57BL/6 (B6) substrains (C57BL/6NJ (B6NJ) and C57BL/6J (B6J)) that differ in their innate neuroimmune response. Herein, we determined the efficacy of A33 for reducing MA self-administration and MA-seeking behavior in these two B6 substrains. Female and male mice of both substrains were first trained to nose poke for a 100 mg/L MA solution followed by a characterization of the dose-response function for oral MA reinforcement (20 mg/L-3.2 g/L), the demand-response function for 400 mg/L MA, and cue-elicited MA seeking following a period of forced abstinence. During this substrain comparison of MA self-administration, we also determined the dose-response function for A33 pretreatment (0-1 mg/kg) on the maintenance of MA self-administration and cue-elicited MA seeking. Relative to B6NJ mice, B6J mice earned fewer reinforcers, consumed less MA, and took longer to reach acquisition criterion with males of both substrains exhibiting some signs of lower MA reinforcement than their female counterparts during the acquisition phase of the study. A33 pretreatment reduced MA reinforcement at all doses tested. These findings provide the first evidence that pretreatment with a selective PDE4B inhibitor effectively reduces MA self-administration in both male and female mice of two genetically distinct substrains but does not impact cue-elicited MA seeking following abstinence. If relevant to humans, these results posit the potential clinical utility of A33 or other selective PDE4B inhibitors for curbing active drug-taking in MA use disorder.
Collapse
Affiliation(s)
- Kevin M. Honeywell
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA; (K.M.H.); (E.V.D.)
| | - Eliyana Van Doren
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA; (K.M.H.); (E.V.D.)
| | - Karen K. Szumlinski
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA; (K.M.H.); (E.V.D.)
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA
| |
Collapse
|
21
|
Manigandan S, Yun JW. Loss of cytoplasmic FMR1-interacting protein 2 (CYFIP2) induces browning in 3T3-L1 adipocytes via repression of GABA-BR and activation of mTORC1. J Cell Biochem 2022; 123:863-877. [PMID: 35233844 DOI: 10.1002/jcb.30231] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/05/2022] [Accepted: 02/13/2022] [Indexed: 11/11/2022]
Abstract
Obesity and related metabolic disorders are epidemic diseases. Promoting thermogenesis and a functional increase in the browning of white adipocytes may counteract obesity. On the other hand, the molecular mechanism that regulates brown and beige fat-mediated thermogenesis is unclear. This article reports a molecular network led by cytoplasmic FMR1-interacting protein 2 (CYFIP2) that negatively regulates adipocyte browning in white adipocytes. Although the function of CYFIP2 in Fragile X Syndrome (FXS) and autism have been reported, its physiological roles in adipocytes remain elusive. Therefore, this study examined the physiological consequences of its deprivation in cultured 3T3-L1 white adipocytes using loss-of-function studies. Combined real-time quantitative reverse-transcription polymerase chain reaction and immunoblot analysis showed that the loss of CYFIP2 induces fat browning, as evidenced by the gene and protein expression levels of the brown fat-associated markers. A deficiency of CYFIP2 promoted mitochondrial biogenesis and significantly enhanced the expression of the core set beige fat-specific genes (Cd137, Cidea, Cited1, Tbx1, and Tmem26) and proteins (PGC-1α, PRDM16, and UCP1). In addition, a CYFIP2 deficiency promoted lipid catabolism and suppressed adipogenesis, lipogenesis, and autophagy. A mechanistic study showed that the loss of CYFIP2 induces browning in white adipocytes, independently via the activation of mTORC1 and suppression of the GABA-BR signaling pathway. The present data revealed a previously unidentified mechanism of CYFIP2 in the browning of white adipocytes and emphasized the potential of CYFIP2 as a pharmacotherapeutic target for treating obesity and other metabolic disorders.
Collapse
Affiliation(s)
- Subramani Manigandan
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, Republic of Korea
| | - Jong Won Yun
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, Republic of Korea
| |
Collapse
|
22
|
Beierle JA, Yao EJ, Goldstein SI, Scotellaro JL, Sena KD, Linnertz CA, Willits AB, Kader L, Young EE, Peltz G, Emili A, Ferris MT, Bryant CD. Genetic basis of thermal nociceptive sensitivity and brain weight in a BALB/c reduced complexity cross. Mol Pain 2022; 18:17448069221079540. [PMID: 35088629 PMCID: PMC8891926 DOI: 10.1177/17448069221079540] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/20/2022] [Indexed: 11/30/2022] Open
Abstract
Thermal nociception involves the transmission of temperature-related noxious information from the periphery to the CNS and is a heritable trait that could predict transition to persistent pain. Rodent forward genetics complement human studies by controlling genetic complexity and environmental factors, analysis of end point tissue, and validation of variants on appropriate genetic backgrounds. Reduced complexity crosses between nearly identical inbred substrains with robust trait differences can greatly facilitate unbiased discovery of novel genes and variants. We found BALB/cByJ mice showed enhanced sensitivity on the 53.5°C hot plate and mechanical stimulation in the von Frey test compared to BALB/cJ mice and replicated decreased gross brain weight in BALB/cByJ versus BALB/cJ. We then identified a quantitative trait locus (QTL) on chromosome 13 for hot plate sensitivity (LOD = 10.7; p < 0.001; peak = 56 Mb) and a QTL for brain weight on chromosome 5 (LOD = 8.7; p < 0.001). Expression QTL mapping of brain tissues identified H2afy (56.07 Mb) as the top transcript with the strongest association at the hot plate locus (FDR = 0.0002) and spliceome analysis identified differential exon usage within H2afy associated with the same locus. Whole brain proteomics further supported decreased H2AFY expression could underlie enhanced hot plate sensitivity, and identified ACADS as a candidate for reduced brain weight. To summarize, a BALB/c reduced complexity cross combined with multiple-omics approaches facilitated identification of candidate genes underlying thermal nociception and brain weight. These substrains provide a powerful, reciprocal platform for future validation of candidate variants.
Collapse
Affiliation(s)
- Jacob A Beierle
- Program in Biomolecular Pharmacology, Boston University School of Medicine, Boston, MA, USA
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Emily J Yao
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Stanley I Goldstein
- Program in Biomolecular Pharmacology, Boston University School of Medicine, Boston, MA, USA
- Department of Biology and Biochemistry, Center for Network Systems Biology, Boston University School of Medicine, Boston, MA, USA
| | - Julia L Scotellaro
- Department of Biology and Biochemistry, Center for Network Systems Biology, Boston University School of Medicine, Boston, MA, USA
- Undergraduate Research Opportunity Program, Boston University, Boston, MA, USA
| | - Katherine D Sena
- Department of Biology and Biochemistry, Center for Network Systems Biology, Boston University School of Medicine, Boston, MA, USA
- Undergraduate Research Opportunity Program, Boston University, Boston, MA, USA
| | - Colton A Linnertz
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adam B Willits
- Neuroscience Program, University of Kansas Medical Center, Kansas City, KS, USA
| | - Leena Kader
- Neuroscience Program, University of Kansas Medical Center, Kansas City, KS, USA
| | - Erin E Young
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Gary Peltz
- Department of Anesthesiology, Pain, and Preoperative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Andrew Emili
- Department of Biology and Biochemistry, Center for Network Systems Biology, Boston University School of Medicine, Boston, MA, USA
| | - Martin T Ferris
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Camron D Bryant
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
23
|
Miura I, Kikkawa Y, Yasuda SP, Shinogi A, Usuda D, Kumar V, Takahashi JS, Tamura M, Masuya H, Wakana S. Characterization of single nucleotide polymorphisms for a forward genetics approach using genetic crosses in C57BL/6 and BALB/c substrains of mice. Exp Anim 2021; 71:240-251. [PMID: 34980769 PMCID: PMC9130033 DOI: 10.1538/expanim.21-0181] [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] [Indexed: 11/20/2022] Open
Abstract
Forward genetics is a powerful approach based on chromosomal mapping of phenotypes and has successfully led to the discovery of many mouse mutations in genes responsible for various
phenotypes. Although crossing between genetically remote strains can produce F2 and backcross mice for chromosomal mapping, the phenotypes are often affected by background effects
from the partner strains in genetic crosses. Genetic crosses between substrains might be useful in genetic mapping to avoid genetic background effects. In this study, we investigated single
nucleotide polymorphisms (SNPs) available for genetic mapping using substrains of C57BL/6 and BALB/c mice. In C57BL/6 mice, 114 SNP markers were developed and assigned to locations on all
chromosomes for full utilization for genetic mapping using genetic crosses between the C57BL/6J and C57BL/6N substrains. Moreover, genetic differences were identified in the 114 SNP markers
among the seven C57BL/6 substrains from five production breeders. In addition, 106 SNPs were detected on all chromosomes of BALB/cAJcl and BALB/cByJJcl substrains. These SNPs could be used
for genotyping in BALB/cJ, BALB/cAJcl, BALB/cAnNCrlCrlj, and BALB/cCrSlc mice, and they are particularly useful for genetic mapping using crosses between BALB/cByJJcl and other BALB/c
substrains. The SNPs characterized in this study can be utilized for genetic mapping to identify the causative mutations of the phenotypes induced by N-ethyl-N-nitrosourea mutagenesis and
the SNPs responsible for phenotypic differences between the substrains of C57BL/6 and BALB/c mice.
Collapse
Affiliation(s)
- Ikuo Miura
- Division of Molecular Genetics, Department of Cooperative Graduate School, School of Medicine, Faculty of Medicine, Graduate School of Medical and Dental Sciences (Medicine), Niigata University.,Technology and Development Team for Mouse Phenotype Analysis, Japan Mouse Clinic, RIKEN BioResource Research Center.,Deafness Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science
| | - Yoshiaki Kikkawa
- Division of Molecular Genetics, Department of Cooperative Graduate School, School of Medicine, Faculty of Medicine, Graduate School of Medical and Dental Sciences (Medicine), Niigata University.,Deafness Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science
| | - Shumpei P Yasuda
- Deafness Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science
| | - Akiko Shinogi
- Technology and Development Team for Mouse Phenotype Analysis, Japan Mouse Clinic, RIKEN BioResource Research Center
| | - Daiki Usuda
- Technology and Development Team for Mouse Phenotype Analysis, Japan Mouse Clinic, RIKEN BioResource Research Center.,Integrated Bioresource Information Division, RIKEN BioResource Research Center
| | | | - Joseph S Takahashi
- Department of Neuroscience, University of Texas Southwestern Medical Center
| | - Masaru Tamura
- Technology and Development Team for Mouse Phenotype Analysis, Japan Mouse Clinic, RIKEN BioResource Research Center
| | - Hiroshi Masuya
- Integrated Bioresource Information Division, RIKEN BioResource Research Center
| | - Shigeharu Wakana
- Technology and Development Team for Mouse Phenotype Analysis, Japan Mouse Clinic, RIKEN BioResource Research Center.,Department of Animal Experimentation, Foundation for Biomedical Research and Innovation at Kobe, Creative Lab for Innovation in Kobe
| |
Collapse
|
24
|
Rehn S, Raymond JS, Boakes RA, Leenaars CHC. A systematic review and meta-analysis of animal models of binge eating - Part 1: Definitions and food/drink intake outcomes. Neurosci Biobehav Rev 2021; 132:1137-1156. [PMID: 34742923 DOI: 10.1016/j.neubiorev.2021.10.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/19/2021] [Accepted: 10/28/2021] [Indexed: 12/09/2022]
Abstract
Binge eating involves consuming excessive amounts of food within a discrete period of time and is associated with significant impairments in binge-eating disorder and bulimia nervosa. While research on clinical binge eating has provided valuable aetiological insights, animal models allow for closer examination of environmental, biological, and developmental risk factors. Numerous animal models of binge eating exist and differ widely in operational definitions of bingeing, animal characteristics and methodological parameters. The current review aimed to synthesise the available published evidence on these models. A systematic review of binge definitions in 170 articles found most studies displayed good face validity. Meta-analyses on 150 articles confirmed that the amount of food or drink consumed by animals under binge conditions was larger than that of non-binge conditions across many protocols. The meta-regression revealed species, strain, and sex moderated binge effect size, with the largest effect observed in studies with female animals and mice. Risk of bias assessment identified that improved reporting of allocation, baseline characteristics and outcome assessment is required in future studies.
Collapse
Affiliation(s)
- Simone Rehn
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, 2006 NSW, Australia.
| | - Joel S Raymond
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, 2006 NSW, Australia; The University of Sydney, Brain and Mind Centre, 94 Mallett Street, Camperdown, Sydney, NSW 2050, Australia
| | - Robert A Boakes
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, 2006 NSW, Australia
| | - Cathalijn H C Leenaars
- Institute for Laboratory Animal Science, Hannover Medical School, 30625, Hannover, Germany; Department for Health Evidence, Radboud University Medical Centre, 6600, Nijmegen, The Netherlands
| |
Collapse
|
25
|
Goldberg LR, Yao EJ, Kelliher JC, Reed ER, Cox JW, Parks C, Kirkpatrick SL, Beierle JA, Chen MM, Johnson WE, Homanics GE, Williams RW, Bryant CD, Mulligan MK. A quantitative trait variant in Gabra2 underlies increased methamphetamine stimulant sensitivity. GENES, BRAIN, AND BEHAVIOR 2021; 20:e12774. [PMID: 34677900 PMCID: PMC9083095 DOI: 10.1111/gbb.12774] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/19/2021] [Accepted: 09/15/2021] [Indexed: 12/24/2022]
Abstract
Psychostimulant (methamphetamine, cocaine) use disorders have a genetic component that remains mostly unknown. We conducted genome-wide quantitative trait locus (QTL) analysis of methamphetamine stimulant sensitivity. To facilitate gene identification, we employed a Reduced Complexity Cross between closely related C57BL/6 mouse substrains and examined maximum speed and distance traveled over 30 min following methamphetamine (2 mg/kg, i.p.). For maximum methamphetamine-induced speed following the second and third administration, we identified a single genome-wide significant QTL on chromosome 11 that peaked near the Cyfip2 locus (LOD = 3.5, 4.2; peak = 21 cM [36 Mb]). For methamphetamine-induced distance traveled following the first and second administration, we identified a genome-wide significant QTL on chromosome 5 that peaked near a functional intronic indel in Gabra2 coding for the alpha-2 subunit of the GABA-A receptor (LOD = 3.6-5.2; peak = 34-35 cM [66-67 Mb]). Striatal cis-expression QTL mapping corroborated Gabra2 as a functional candidate gene underlying methamphetamine-induced distance traveled. CRISPR/Cas9-mediated correction of the mutant intronic deletion on the C57BL/6J background to the wild-type C57BL/6NJ allele was sufficient to reduce methamphetamine-induced locomotor activity toward the wild-type C57BL/6NJ-like level, thus validating the quantitative trait variant (QTV). These studies show the power and efficiency of Reduced Complexity Crosses in identifying causal variants underlying complex traits. Functionally restoring Gabra2 expression decreased methamphetamine stimulant sensitivity and supports preclinical and human genetic studies implicating the GABA-A receptor in psychostimulant addiction-relevant traits. Importantly, our findings have major implications for studying psychostimulants in the C57BL/6J strain-the gold standard strain in biomedical research.
Collapse
Affiliation(s)
- Lisa R. Goldberg
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston, Massachusetts, USA
- NIGMS T32 Ph.D. Training Program in Biomolecular Pharmacology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Emily J. Yao
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston, Massachusetts, USA
| | - Julia C. Kelliher
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston, Massachusetts, USA
| | - Eric R. Reed
- Ph.D. Program in Bioinformatics, Boston University, Boston, Massachusetts, USA
| | - Jiayi Wu Cox
- Program in Biomedical Sciences, Graduate Program in Genetics and Genomics, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Cory Parks
- Department of Agricultural, Biology, and Health Sciences, Cameron University, Lawton, Oklahoma, USA
| | - Stacey L. Kirkpatrick
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston, Massachusetts, USA
| | - Jacob A. Beierle
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston, Massachusetts, USA
- NIGMS T32 Ph.D. Training Program in Biomolecular Pharmacology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Melanie M. Chen
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston, Massachusetts, USA
| | - William E. Johnson
- Department of Medicine, Computational Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Gregg E. Homanics
- Departments of Anesthesiology, Neurobiology, and Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert W. Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Camron D. Bryant
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston, Massachusetts, USA
| | - Megan K. Mulligan
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
26
|
Kantak KM, Stots C, Mathieson E, Bryant CD. Spontaneously Hypertensive Rat substrains show differences in model traits for addiction risk and cocaine self-administration: Implications for a novel rat reduced complexity cross. Behav Brain Res 2021; 411:113406. [PMID: 34097899 PMCID: PMC8265396 DOI: 10.1016/j.bbr.2021.113406] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/28/2021] [Accepted: 06/03/2021] [Indexed: 12/17/2022]
Abstract
Forward genetic mapping of F2 crosses between closely related substrains of inbred rodents - referred to as a reduced complexity cross (RCC) - is a relatively new strategy for accelerating the pace of gene discovery for complex traits, such as drug addiction. RCCs to date were generated in mice, but rats are thought to be optimal for addiction genetic studies. Based on past literature, one inbred Spontaneously Hypertensive Rat substrain, SHR/NCrl, is predicted to exhibit a distinct behavioral profile as it relates to cocaine self-administration traits relative to another substrain, SHR/NHsd. Direct substrain comparisons are a necessary first step before implementing an RCC. We evaluated model traits for cocaine addiction risk and cocaine self-administration behaviors using a longitudinal within-subjects design. Impulsive-like and compulsive-like traits were greater in SHR/NCrl than SHR/NHsd, as were reactivity to sucrose reward, sensitivity to acute psychostimulant effects of cocaine, and cocaine use studied under fixed-ratio and tandem schedules of cocaine self-administration. Compulsive-like behavior correlated with the acute psychostimulant effects of cocaine, which in turn correlated with cocaine taking under the tandem schedule. Compulsive-like behavior also was the best predictor of cocaine seeking responses. Heritability estimates indicated that 22 %-40 % of the variances for the above phenotypes can be explained by additive genetic factors, providing sufficient genetic variance to conduct genetic mapping in F2 crosses of SHR/NCrl and SHR/NHsd. These results provide compelling support for using an RCC approach in SHR substrains to uncover candidate genes and variants that are of relevance to cocaine use disorders.
Collapse
Affiliation(s)
- Kathleen M Kantak
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA; Center for Systems Neuroscience, Boston University, Boston, MA, USA.
| | - Carissa Stots
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA
| | - Elon Mathieson
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA
| | - Camron D Bryant
- Departments of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA; Center for Systems Neuroscience, Boston University, Boston, MA, USA
| |
Collapse
|
27
|
Biembengut ÍV, Silva ILZ, Souza TDACBD, Shigunov P. Cytoplasmic FMR1 interacting protein (CYFIP) family members and their function in neural development and disorders. Mol Biol Rep 2021; 48:6131-6143. [PMID: 34327661 DOI: 10.1007/s11033-021-06585-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/20/2021] [Indexed: 11/25/2022]
Abstract
In humans, the cytoplasmic FMR1 interacting protein (CYFIP) family is composed of CYFIP1 and CYFIP2. Despite their high similarity and shared interaction with many partners, CYFIP1 and CYFIP2 act at different points in cellular processes. CYFIP1 and CYFIP2 have different expression levels in human tissues, and knockout animals die at different time points of development. CYFIP1, similar to CYFIP2, acts in the WAVE regulatory complex (WRC) and plays a role in actin dynamics through the activation of the Arp2/3 complex and in a posttranscriptional regulatory complex with the fragile X mental retardation protein (FMRP). Previous reports have shown that CYFIP1 and CYFIP2 may play roles in posttranscriptional regulation in different ways. While CYFIP1 is involved in translation initiation via the 5'UTR, CYFIP2 may regulate mRNA expression via the 3'UTR. In addition, this CYFIP protein family is involved in neural development and maturation as well as in different neural disorders, such as intellectual disabilities, autistic spectrum disorders, and Alzheimer's disease. In this review, we map diverse studies regarding the functions, regulation, and implications of CYFIP proteins in a series of molecular pathways. We also highlight mutations and their structural effects both in functional studies and in neural diseases.
Collapse
Affiliation(s)
- Ísis Venturi Biembengut
- Carlos Chagas Institute-FIOCRUZ-PR, Rua Prof. Algacyr Munhoz Mader, 3775, CIC, Curitiba, Paraná, 81830-010, Brazil
| | | | | | - Patrícia Shigunov
- Carlos Chagas Institute-FIOCRUZ-PR, Rua Prof. Algacyr Munhoz Mader, 3775, CIC, Curitiba, Paraná, 81830-010, Brazil.
| |
Collapse
|
28
|
Jimenez Chavez CL, Bryant CD, Munn-Chernoff MA, Szumlinski KK. Selective Inhibition of PDE4B Reduces Binge Drinking in Two C57BL/6 Substrains. Int J Mol Sci 2021; 22:ijms22115443. [PMID: 34064099 PMCID: PMC8196757 DOI: 10.3390/ijms22115443] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/16/2021] [Accepted: 05/18/2021] [Indexed: 01/15/2023] Open
Abstract
Cyclic AMP (cAMP)-dependent signaling is highly implicated in the pathophysiology of alcohol use disorder (AUD), with evidence supporting the efficacy of inhibiting the cAMP hydrolyzing enzyme phosphodiesterase 4 (PDE4) as a therapeutic strategy for drinking reduction. Off-target emetic effects associated with non-selective PDE4 inhibitors has prompted the development of selective PDE4 isozyme inhibitors for treating neuropsychiatric conditions. Herein, we examined the effect of a selective PDE4B inhibitor A33 (0–1.0 mg/kg) on alcohol drinking in both female and male mice from two genetically distinct C57BL/6 substrains. Under two different binge-drinking procedures, A33 pretreatment reduced alcohol intake in male and female mice of both substrains. In both drinking studies, there was no evidence for carry-over effects the next day; however, we did observe some sign of tolerance to A33’s effect on alcohol intake upon repeated, intermittent, treatment (5 injections of 1.0 mg/kg, every other day). Pretreatment with 1.0 mg/kg of A33 augmented sucrose intake by C57BL/6NJ, but not C57BL/6J, mice. In mice with a prior history of A33 pretreatment during alcohol-drinking, A33 (1.0 mg/kg) did not alter spontaneous locomotor activity or basal motor coordination, nor did it alter alcohol’s effects on motor activity, coordination or sedation. In a distinct cohort of alcohol-naïve mice, acute pretreatment with 1.0 mg/kg of A33 did not alter motor performance on a rotarod and reduced sensitivity to the acute intoxicating effects of alcohol. These data provide the first evidence that selective PDE4B inhibition is an effective strategy for reducing excessive alcohol intake in murine models of binge drinking, with minimal off-target effects. Despite reducing sensitivity to acute alcohol intoxication, PDE4B inhibition reduces binge alcohol drinking, without influencing behavioral sensitivity to alcohol in alcohol-experienced mice. Furthermore, A33 is equally effective in males and females and exerts a quantitatively similar reduction in alcohol intake in mice with a genetic predisposition for high versus moderate alcohol preference. Such findings further support the safety and potential clinical utility of targeting PDE4 for treating AUD.
Collapse
Affiliation(s)
- C. Leonardo Jimenez Chavez
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA;
| | - Camron D. Bryant
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA 02118, USA;
| | - Melissa A. Munn-Chernoff
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Karen K. Szumlinski
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA;
- Correspondence:
| |
Collapse
|
29
|
Yao EJ, Babbs RK, Kelliher JC, Luttik KP, Borrelli KN, Damaj MI, Mulligan MK, Bryant CD. Systems genetic analysis of binge-like eating in a C57BL/6J x DBA/2J-F2 cross. GENES, BRAIN, AND BEHAVIOR 2021; 20:e12751. [PMID: 33978997 PMCID: PMC9361732 DOI: 10.1111/gbb.12751] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 05/04/2021] [Accepted: 05/11/2021] [Indexed: 12/19/2022]
Abstract
Binge eating is a heritable trait associated with eating disorders and refers to the rapid consumption of a large quantity of energy-dense food that is, associated with loss of control and negative affect. Binge eating disorder is the most common eating disorder in the United States; however, the genetic basis is unknown. We previously identified robust mouse inbred strain differences between C57BL/6J and DBA/2J in binge-like eating of sweetened palatable food in an intermittent access, conditioned place preference paradigm. To map the genetic basis of changes in body weight and binge-like eating (BLE) and to identify candidate genes, we conducted quantitative trait locus (QTL) analysis in 128 C57BL/6J x DBA/2J-F2 mice combined with PheQTL and trait covariance analysis in GeneNetwork2 using legacy BXD-RI trait datasets. We identified a QTL on Chromosome 18 influencing changes in body weight across days in females (log of the odds [LOD] = 6.3; 1.5-LOD: 3-12 cM) that contains the candidate gene Zeb1. We also identified a sex-combined QTL influencing initial palatable food intake on Chromosome 5 (LOD = 5.8; 1.5-LOD: 21-28 cM) that contains the candidate gene Lcorl and a second QTL influencing escalated palatable food intake on Chromosome 6 in males (LOD = 5.4; 1.5-LOD: 50-59 cM) that contains the candidate genes Adipor2 and Plxnd1. Finally, we identified a suggestive QTL in females for slope of BLE on distal Chromosome 18 (LOD = 4.1; p = 0.055; 1.5-LOD: 23-35 cM). Future studies will use BXD-RI strains to fine map loci and support candidate gene nomination for gene editing.
Collapse
Affiliation(s)
- Emily J. Yao
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA 02118 USA
| | - Richard K. Babbs
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA 02118 USA
| | - Julia C. Kelliher
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA 02118 USA
| | - Kimberly P. Luttik
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA 02118 USA
| | - Kristyn N. Borrelli
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA 02118 USA
- Graduate Program for Neuroscience, Boston University, Boston, MA 02215 USA
- Tranformative Training Program in Addiction Science (TTPAS), Boston University, Boston, MA 02118 USA
- Biomolecluar Pharmacology Training Program, Boston University School of Medicine, Boston, MA 02118 USA
| | - M. Imad Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Megan K. Mulligan
- Department of Genetics, Genomics, and Informatics, The University of Tennessee Health Science Center, Memphis, TN 38163 USA
| | - Camron D. Bryant
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA 02118 USA
- Graduate Program for Neuroscience, Boston University, Boston, MA 02215 USA
- Tranformative Training Program in Addiction Science (TTPAS), Boston University, Boston, MA 02118 USA
- Biomolecluar Pharmacology Training Program, Boston University School of Medicine, Boston, MA 02118 USA
| |
Collapse
|
30
|
Abstract
The inbred mouse strain C57BL/6 has been widely used as a background strain for spontaneous and induced mutations. Developed in the 1930s, the C57BL/6 strain
diverged into two major groups in the 1950s, namely, C57BL/6J and C57BL/6N, and more than 20 substrains have been established from them worldwide. We previously
reported genetic differences among C57BL/6 substrains in 2009 and 2015. Since then, dozens of reports have been published on phenotypic differences in
behavioral, neurological, cardiovascular, and metabolic traits. Substrains need to be chosen according to the purpose of the study because phenotypic
differences might affect the experimental results. In this paper, we review recent reports of phenotypic and genetic differences among C57BL/6 substrains, focus
our attention on the proper use of C57BL/6 and other inbred strains in the era of genome editing, and provide the life science research community wider
knowledge about this subject.
Collapse
Affiliation(s)
- Kazuyuki Mekada
- Department of Zoology, Okayama University of Science, 1-1 Ridai-cho, Kita-ku, Okayama 700-0005, Japan.,Experimental Animal Division, RIKEN BioResource Research Center, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Atsushi Yoshiki
- Experimental Animal Division, RIKEN BioResource Research Center, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| |
Collapse
|
31
|
Salcedo-Arellano MJ, Cabal-Herrera AM, Punatar RH, Clark CJ, Romney CA, Hagerman RJ. Overlapping Molecular Pathways Leading to Autism Spectrum Disorders, Fragile X Syndrome, and Targeted Treatments. Neurotherapeutics 2021; 18:265-283. [PMID: 33215285 PMCID: PMC8116395 DOI: 10.1007/s13311-020-00968-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorders (ASD) are subdivided into idiopathic (unknown) etiology and secondary, based on known etiology. There are hundreds of causes of ASD and most of them are genetic in origin or related to the interplay of genetic etiology and environmental toxicology. Approximately 30 to 50% of the etiologies can be identified when using a combination of available genetic testing. Many of these gene mutations are either core components of the Wnt signaling pathway or their modulators. The full mutation of the fragile X mental retardation 1 (FMR1) gene leads to fragile X syndrome (FXS), the most common cause of monogenic origin of ASD, accounting for ~ 2% of the cases. There is an overlap of molecular mechanisms in those with idiopathic ASD and those with FXS, an interaction between various signaling pathways is suggested during the development of the autistic brain. This review summarizes the cross talk between neurobiological pathways found in ASD and FXS. These signaling pathways are currently under evaluation to target specific treatments in search of the reversal of the molecular abnormalities found in both idiopathic ASD and FXS.
Collapse
Affiliation(s)
- Maria Jimena Salcedo-Arellano
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA.
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA, 95817, USA.
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA.
| | - Ana Maria Cabal-Herrera
- Group on Congenital Malformations and Dysmorphology, Faculty of Health, Universidad del Valle, Cali, 00000, Colombia
| | - Ruchi Harendra Punatar
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA
| | - Courtney Jessica Clark
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA
| | - Christopher Allen Romney
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA
| | - Randi J Hagerman
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA.
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA.
| |
Collapse
|
32
|
Petrick HL, Foley KP, Zlitni S, Brunetta HS, Paglialunga S, Miotto PM, Politis-Barber V, O’Dwyer C, Philbrick DJ, Fullerton MD, Schertzer JD, Holloway GP. Adipose Tissue Inflammation Is Directly Linked to Obesity-Induced Insulin Resistance, while Gut Dysbiosis and Mitochondrial Dysfunction Are Not Required. FUNCTION (OXFORD, ENGLAND) 2020; 1:zqaa013. [PMID: 34278304 PMCID: PMC8276887 DOI: 10.1093/function/zqaa013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/17/2020] [Accepted: 08/21/2020] [Indexed: 01/06/2023]
Abstract
Obesity is associated with adipose tissue hypertrophy, systemic inflammation, mitochondrial dysfunction, and intestinal dysbiosis. Rodent models of high-fat diet (HFD)-feeding or genetic deletion of multifunctional proteins involved in immunity and metabolism are often used to probe the etiology of obesity; however, these models make it difficult to divorce the effects of obesity, diet composition, or immunity on endocrine regulation of blood glucose. We, therefore, investigated the importance of adipose inflammation, mitochondrial dysfunction, and gut dysbiosis for obesity-induced insulin resistance using a spontaneously obese mouse model. We examined metabolic changes in skeletal muscle, adipose tissue, liver, the intestinal microbiome, and whole-body glucose control in spontaneously hyperphagic C57Bl/6J mice compared to lean littermates. A separate subset of lean and obese mice was subject to 8 weeks of obesogenic HFD feeding, or to pair feeding of a standard rodent diet. Hyperphagia, obesity, adipose inflammation, and insulin resistance were present in obese mice despite consuming a standard rodent diet, and these effects were blunted with caloric restriction. However, hyperphagic obese mice had normal mitochondrial respiratory function in all tissues tested and no discernable intestinal dysbiosis relative to lean littermates. In contrast, feeding mice an obesogenic HFD altered the composition of the gut microbiome, impaired skeletal muscle mitochondrial bioenergetics, and promoted poor glucose control. These data show that adipose inflammation and redox stress occurred in all models of obesity, but gut dysbiosis and mitochondrial respiratory dysfunction are not always required for obesity-induced insulin resistance. Rather, changes in the intestinal microbiome and mitochondrial bioenergetics may reflect physiological consequences of HFD feeding.
Collapse
Affiliation(s)
- Heather L Petrick
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Kevin P Foley
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Soumaya Zlitni
- Departments of Genetics and Medicine, Stanford University, Stanford, 94305, CA, USA
| | - Henver S Brunetta
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada,Department of Physiological Sciences, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Sabina Paglialunga
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Paula M Miotto
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Valerie Politis-Barber
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Conor O’Dwyer
- Department of Biochemistry, Microbiology and Immunology, Centre for Inflammation, Infection and Immunity, Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON, Canada
| | - Diana J Philbrick
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Morgan D Fullerton
- Department of Biochemistry, Microbiology and Immunology, Centre for Inflammation, Infection and Immunity, Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON, Canada
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Graham P Holloway
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada,Address correspondence to G.P.H. (e-mail: )
| |
Collapse
|
33
|
Facilitating Complex Trait Analysis via Reduced Complexity Crosses. Trends Genet 2020; 36:549-562. [PMID: 32482413 DOI: 10.1016/j.tig.2020.05.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 05/05/2020] [Accepted: 05/12/2020] [Indexed: 01/02/2023]
Abstract
Genetically diverse inbred strains are frequently used in quantitative trait mapping to identify sequence variants underlying trait variation. Poor locus resolution and high genetic complexity impede variant discovery. As a solution, we explore reduced complexity crosses (RCCs) between phenotypically divergent, yet genetically similar, rodent substrains. RCCs accelerate functional variant discovery via decreasing the number of segregating variants by orders of magnitude. The simplified genetic architecture of RCCs often permit immediate identification of causal variants or rapid fine-mapping of broad loci to smaller intervals. Whole-genome sequences of substrains make RCCs possible by supporting the development of array- and targeted sequencing-based genotyping platforms, coupled with rapid genome editing for variant validation. In summary, RCCs enhance discovery-based genetics of complex traits.
Collapse
|
34
|
Gondré-Lewis MC, Bassey R, Blum K. Pre-clinical models of reward deficiency syndrome: A behavioral octopus. Neurosci Biobehav Rev 2020; 115:164-188. [PMID: 32360413 DOI: 10.1016/j.neubiorev.2020.04.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 04/08/2020] [Accepted: 04/20/2020] [Indexed: 12/15/2022]
Abstract
Individuals with mood disorders or with addiction, impulsivity and some personality disorders can share in common a dysfunction in how the brain perceives reward, where processing of natural endorphins or the response to exogenous dopamine stimulants is impaired. Reward Deficiency Syndrome (RDS) is a polygenic trait with implications that suggest cross-talk between different neurological systems that include the known reward pathway, neuroendocrine systems, and motivational systems. In this review we evaluate well-characterized animal models for their construct validity and as potential models for RDS. Animal models used to study substance use disorder, major depressive disorder (MDD), early life stress, immune dysregulation, attention deficit hyperactivity disorder (ADHD), post traumatic stress disorder (PTSD), compulsive gambling and compulsive eating disorders are discussed. These disorders recruit underlying reward deficiency mechanisms in multiple brain centers. Because of the widespread and remarkable array of associated/overlapping behavioral manifestations with a common root of hypodopaminergia, the basic endophenotype recognized as RDS is indeed likened to a behavioral octopus. We conclude this review with a look ahead on how these models can be used to investigate potential therapeutics that target the underlying common deficiency.
Collapse
Affiliation(s)
- Marjorie C Gondré-Lewis
- Department of Anatomy, Howard University College of Medicine, 520 W Street, NW, Washington D.C., 20059, United States; Developmental Neuropsychopharmacology Laboratory, Howard University College of Medicine, 520 W Street, NW, Washington D.C., 20059, United States.
| | - Rosemary Bassey
- Developmental Neuropsychopharmacology Laboratory, Howard University College of Medicine, 520 W Street, NW, Washington D.C., 20059, United States; Department of Science Education, Donald and Barbara Zucker School of Medicine at Hofstra/ Northwell, 500 Hofstra University, Hempstead, NY 11549, United States
| | - Kenneth Blum
- Western University Health Sciences, Graduate College of Biomedical Sciences, Pomona, California, United States
| |
Collapse
|
35
|
Davenport EC, Szulc BR, Drew J, Taylor J, Morgan T, Higgs NF, López-Doménech G, Kittler JT. Autism and Schizophrenia-Associated CYFIP1 Regulates the Balance of Synaptic Excitation and Inhibition. Cell Rep 2020; 26:2037-2051.e6. [PMID: 30784587 PMCID: PMC6381785 DOI: 10.1016/j.celrep.2019.01.092] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 10/26/2018] [Accepted: 01/24/2019] [Indexed: 12/28/2022] Open
Abstract
Altered excitatory/inhibitory (E/I) balance is implicated in neuropsychiatric and neurodevelopmental disorders, but the underlying genetic etiology remains poorly understood. Copy number variations in CYFIP1 are associated with autism, schizophrenia, and intellectual disability, but its role in regulating synaptic inhibition or E/I balance remains unclear. We show that CYFIP1, and the paralog CYFIP2, are enriched at inhibitory postsynaptic sites. While CYFIP1 or CYFIP2 upregulation increases excitatory synapse number and the frequency of miniature excitatory postsynaptic currents (mEPSCs), it has the opposite effect at inhibitory synapses, decreasing their size and the amplitude of miniature inhibitory postsynaptic currents (mIPSCs). Contrary to CYFIP1 upregulation, its loss in vivo, upon conditional knockout in neocortical principal cells, increases expression of postsynaptic GABAA receptor β2/3-subunits and neuroligin 3, enhancing synaptic inhibition. Thus, CYFIP1 dosage can bi-directionally impact inhibitory synaptic structure and function, potentially leading to altered E/I balance and circuit dysfunction in CYFIP1-associated neurological disorders. CYFIP1 and CYFIP2 are enriched at inhibitory synapses. CYFIP1 upregulation differentially disrupts inhibitory and excitatory synapses. Conditional loss of CYFIP1 alters neuroligin 3 and GABAAR β-subunits expression. Loss of CYFIP1 increases inhibitory synaptic clusters and hence mIPSC amplitude.
Collapse
Affiliation(s)
- Elizabeth C Davenport
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Blanka R Szulc
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - James Drew
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - James Taylor
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Toby Morgan
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Nathalie F Higgs
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Guillermo López-Doménech
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Josef T Kittler
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
36
|
Babbs RK, Beierle JA, Yao EJ, Kelliher JC, Medeiros AR, Anandakumar J, Shah AA, Chen MM, Johnson WE, Bryant CD. The effect of the demyelinating agent cuprizone on binge-like eating of sweetened palatable food in female and male C57BL/6 substrains. Appetite 2020; 150:104678. [PMID: 32209386 DOI: 10.1016/j.appet.2020.104678] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/26/2020] [Accepted: 03/18/2020] [Indexed: 12/16/2022]
Abstract
Binge eating is a heritable symptom of eating disorders with an unknown genetic etiology. Rodent models for binge-like eating (BLE) of palatable food permit the study of genetic and biological mechanisms. We previously genetically mapped a coding mutation in Cyfip2 associated with increased BLE of sweetened palatable food in the C57BL/6NJ versus C57BL/6J substrain. The increase in BLE in C57BL/6NJ mice was associated with a decrease in transcription of genes enriched for myelination in the striatum. Here, we tested the hypothesis that decreasing myelin levels with the demyelinating agent cuprizone would enhance BLE. Mice were treated with a 0.3% cuprizone home cage diet for two weeks. Cuprizone induced similar weight loss in both substrains and sexes that recovered within 48 h after removal of cuprizone. Following a three-week recovery period, mice were trained for BLE in an intermittent, limited access procedure. Surprisingly, cuprizone significantly reduced BLE in male but not female C57BL/6NJ mice while having no effect in C57BL/6J mice. Cuprizone also reduced myelin basic protein (MBP) at seven weeks post-cuprizone removal while having no effect on myelin-associated glycoprotein at this time point. C57BL/6NJ mice also showed less MBP than C57BL/6J mice. There were no statistical interactions of Treatment with Sex on MBP levels, indicating that differences in MBP reduction are unlikely to account for sex differences in BLE. To summarize, cuprizone induced an unexpected, significant reduction in BLE in C57BL/6NJ males, which could indicate genotype-dependent sex differences in the biological mechanisms of BLE.
Collapse
Affiliation(s)
- Richard K Babbs
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, 72 E. Concord St., L-606C, Boston, MA, 02118, USA
| | - Jacob A Beierle
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, 72 E. Concord St., L-606C, Boston, MA, 02118, USA; Biomolecular Pharmacology Ph.D. Program, Boston University School of Medicine, USA; Boston University's Transformative Training Program in Addiction Science (TTPAS), Biomedical Genetics, Boston University School of Medicine, 72 E. Concord St., E-200, Boston, MA, 02118, USA
| | - Emily J Yao
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, 72 E. Concord St., L-606C, Boston, MA, 02118, USA
| | - Julia C Kelliher
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, 72 E. Concord St., L-606C, Boston, MA, 02118, USA
| | - Arthurine R Medeiros
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, 72 E. Concord St., L-606C, Boston, MA, 02118, USA; National Institute on Drug Abuse Diversity Scholars Program, 6001 Executive Boulevard, Room 3105, MSC 9567, Bethesda, MD, USA, 20892-9567
| | - Jeya Anandakumar
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, 72 E. Concord St., L-606C, Boston, MA, 02118, USA; National Institute on Drug Abuse Diversity Scholars Program, 6001 Executive Boulevard, Room 3105, MSC 9567, Bethesda, MD, USA, 20892-9567
| | - Anyaa A Shah
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, 72 E. Concord St., L-606C, Boston, MA, 02118, USA
| | - Melanie M Chen
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, 72 E. Concord St., L-606C, Boston, MA, 02118, USA
| | - William E Johnson
- Department of Medicine, Division of Computational Biomedicine, Boston University, 72 E. Concord St., E-609, Boston, MA, 02118, USA
| | - Camron D Bryant
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, 72 E. Concord St., L-606C, Boston, MA, 02118, USA.
| |
Collapse
|
37
|
Zhang Y, Lee Y, Han K. Neuronal function and dysfunction of CYFIP2: from actin dynamics to early infantile epileptic encephalopathy. BMB Rep 2019. [PMID: 30982501 PMCID: PMC6549915 DOI: 10.5483/bmbrep.2019.52.5.097] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The cytoplasmic FMR1-interacting protein family (CYFIP1 and CYFIP2) are evolutionarily conserved proteins originally identified as binding partners of the fragile X mental retardation protein (FMRP), a messenger RNA (mRNA)-binding protein whose loss causes the fragile X syndrome. Moreover, CYFIP is a key component of the heteropentameric WAVE regulatory complex (WRC), a critical regulator of neuronal actin dynamics. Therefore, CYFIP may play key roles in regulating both mRNA translation and actin polymerization, which are critically involved in proper neuronal development and function. Nevertheless, compared to CYFIP1, neuronal function and dysfunction of CYFIP2 remain largely unknown, possibly due to the relatively less well established association between CYFIP2 and brain disorders. Despite high amino acid sequence homology between CYFIP1 and CYFIP2, several in vitro and animal model studies have suggested that CYFIP2 has some unique neuronal functions distinct from those of CYFIP1. Furthermore, recent whole-exome sequencing studies identified de novo hot spot variants of CYFIP2 in patients with early infantile epileptic encephalopathy (EIEE), clearly implicating CYFIP2 dysfunction in neurological disorders. In this review, we highlight these recent investigations into the neuronal function and dysfunction of CYFIP2, and also discuss several key questions remaining about this intriguing neuronal protein.
Collapse
Affiliation(s)
- Yinhua Zhang
- Departments of Neuroscience, and Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
| | - Yeunkum Lee
- Departments of Neuroscience, and Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
| | - Kihoon Han
- Departments of Neuroscience, and Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
| |
Collapse
|
38
|
Hui KK, Chen YK, Endo R, Tanaka M. Translation from the Ribosome to the Clinic: Implication in Neurological Disorders and New Perspectives from Recent Advances. Biomolecules 2019; 9:E680. [PMID: 31683805 PMCID: PMC6920867 DOI: 10.3390/biom9110680] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/27/2019] [Accepted: 10/29/2019] [Indexed: 12/12/2022] Open
Abstract
De novo protein synthesis by the ribosome and its multitude of co-factors must occur in a tightly regulated manner to ensure that the correct proteins are produced accurately at the right time and, in some cases, also in the proper location. With novel techniques such as ribosome profiling and cryogenic electron microscopy, our understanding of this basic biological process is better than ever and continues to grow. Concurrently, increasing attention is focused on how translational regulation in the brain may be disrupted during the progression of various neurological disorders. In fact, translational dysregulation is now recognized as the de facto pathogenic cause for some disorders. Novel mechanisms including ribosome stalling, ribosome-associated quality control, and liquid-liquid phase separation are closely linked to translational regulation, and may thus be involved in the pathogenic process. The relationships between translational dysregulation and neurological disorders, as well as the ways through which we may be able to reverse those detrimental effects, will be examined in this review.
Collapse
Affiliation(s)
- Kelvin K Hui
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan.
| | - Yi-Kai Chen
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan.
| | - Ryo Endo
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan.
| | - Motomasa Tanaka
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan.
| |
Collapse
|
39
|
Newmyer BA, Whindleton CM, Srinivasa N, Jones MK, Scott MM. Genetic variation affects binge feeding behavior in female inbred mouse strains. Sci Rep 2019; 9:15709. [PMID: 31673099 PMCID: PMC6823456 DOI: 10.1038/s41598-019-51874-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 10/09/2019] [Indexed: 12/17/2022] Open
Abstract
Identifying genetic variants that regulate binge eating (BE) is critical for understanding the factors that control this behavior and for the development of pharmacological treatment strategies. Although several studies have revealed specific genes capable of affecting BE behavior, less is known about how genetic variation modulates BE. Thus, through a paradigm that promoted binge-like food intake through intermittent access to high calorie diet (HCD), we quantified food-intake in four inbred mouse strains: C57Bl/6J (B6), NOD/LtJ (NOD), 129S1/SvlmJ (S1), and A/J (AJ). We report that genetic variation likely influences the chronic regulation of food intake and the binge-like consumption of a palatable HCD. AJ mice consumed more of both standard chow and HCD than the other three strains tested when both diets were available ad libitum, while S1 mice consumed significantly less HCD than other strains during intermittent HCD access. Behavioral differences were also associated with differential changes in c-Fos immunohistochemistry in brain regions traditionally associated with appetite regulation. Our results identify 129S1/SvlmJ as a strain that exhibits low levels of binge feeding behavior and suggests that this strain could be useful in the investigation of the influence of genetic variation in the control of binge food intake.
Collapse
Affiliation(s)
- Brandon A Newmyer
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Ciarra M Whindleton
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Nandan Srinivasa
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Marieke K Jones
- Health Sciences Library, University of Virginia, Charlottesville, VA, USA
| | - Michael M Scott
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
40
|
Levitsky LI, Kliuchnikova AA, Kuznetsova KG, Karpov DS, Ivanov MV, Pyatnitskiy MA, Kalinina OV, Gorshkov MV, Moshkovskii SA. Adenosine-to-Inosine RNA Editing in Mouse and Human Brain Proteomes. Proteomics 2019; 19:e1900195. [PMID: 31576663 DOI: 10.1002/pmic.201900195] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 09/25/2019] [Indexed: 12/30/2022]
Abstract
Proteogenomics is based on the use of customized genome or RNA sequencing databases for interrogation of shotgun proteomics data in search for proteome-level evidence of genome variations or RNA editing. In this work, the products of adenosine-to-inosine RNA editing in human and murine brain proteomes are identified using publicly available brain proteome LC-MS/MS datasets and an RNA editome database compiled from several sources. After filtering of false-positive results, 20 and 37 sites of editing in proteins belonging to 14 and 32 genes are identified for murine and human brain proteomes, respectively. Eight sites of editing identified with high spectral counts overlapped between human and mouse brain samples. Some of these sites have been previously reported using orthogonal methods, such as α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) glutamate receptors, CYFIP2, coatomer alpha. Also, differential editing between neurons and microglia is demonstrated in this work for some of the proteins from primary murine brain cell cultures. Because many edited sites are still not characterized functionally at the protein level, the results provide a necessary background for their further analysis in normal and diseased cells and tissues using targeted proteomic approaches.
Collapse
Affiliation(s)
- Lev I Levitsky
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center of Chemical Physics, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Anna A Kliuchnikova
- Institute of Biomedical Chemistry, 10 Pogodinskaya st., Moscow, 119121, Russia.,Department of Biochemistry, Pirogov Russian National Research Medical University, 1 Ostrovityanova st., Moscow, 117997, Russia
| | - Ksenia G Kuznetsova
- Institute of Biomedical Chemistry, 10 Pogodinskaya st., Moscow, 119121, Russia
| | - Dmitry S Karpov
- Institute of Biomedical Chemistry, 10 Pogodinskaya st., Moscow, 119121, Russia.,Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Mark V Ivanov
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center of Chemical Physics, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Mikhail A Pyatnitskiy
- Institute of Biomedical Chemistry, 10 Pogodinskaya st., Moscow, 119121, Russia.,Onco Genotest LLC, Moscow, 125047, Russia.,Department of Technologies for Complex System Modelling, National Research University Higher School of Economics, Moscow, 101000, Russia
| | - Olga V Kalinina
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, Saarbrücken, 66123, Germany.,Medical Faculty, Saarland University, Kirrberger Straße, Homburg, 66421, Germany
| | - Mikhail V Gorshkov
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center of Chemical Physics, Russian Academy of Sciences, Moscow, 119991, Russia.,Moscow Institute of Physics and Technology (State University), Dolgoprudny, 141700, Moscow Region, Russia
| | - Sergei A Moshkovskii
- Institute of Biomedical Chemistry, 10 Pogodinskaya st., Moscow, 119121, Russia.,Department of Biochemistry, Pirogov Russian National Research Medical University, 1 Ostrovityanova st., Moscow, 117997, Russia
| |
Collapse
|
41
|
Akinola LS, Mckiver B, Toma W, Zhu AZX, Tyndale RF, Kumar V, Damaj MI. C57BL/6 Substrain Differences in Pharmacological Effects after Acute and Repeated Nicotine Administration. Brain Sci 2019; 9:brainsci9100244. [PMID: 31546627 PMCID: PMC6827359 DOI: 10.3390/brainsci9100244] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/18/2019] [Accepted: 09/19/2019] [Indexed: 01/16/2023] Open
Abstract
Tobacco smoking is the major cause of disability and death in the United States and around the world. In addition, tobacco dependence and addiction express themselves as complex behaviors involving an interplay of genetics, environment, and psychological state. Mouse genetic studies could potentially elucidate the novel genes and/or gene networks regulating various aspects of nicotine dependence. Using the closely related C57BL/6 (B6) mice substrains, recent reports have noted phenotypic differences within C57BL/6J (B6J) and C57BL/6N (B6N) mice for some drugs of abuse: alcohol, opiates, and cocaine. However, the differences in nicotine’s effects have not yet been described in these substrains. We examined the phenotypic differences in these substrains following the acute and repeated administration of nicotine in several pharmacological measures, including locomotion (after acute and repeated exposure), body temperature, nociception, and anxiety-like behaviors. We report substrain differences in the pharmacological effects of acute and repeated nicotine administration in the B6 substrains. Overall, we show enhanced nicotine sensitivity to locomotion, hypothermia, antinociception, and anxiety-like behaviors in the B6J mouse substrain compared to B6N. In the repeated administration paradigm, both the B6N and B6J substrains showed no sensitized locomotor responses after repeated exposure to nicotine at the two doses tested. This study thus provides evidence that the B6 mouse substrains may be useful for genetic studies to elucidate some of the genetic variants involved in tobacco dependence and addiction.
Collapse
Affiliation(s)
- Lois S Akinola
- Department of Pharmacology and Toxicology, and Translational Research Initiative for Pain and Neuropathy, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA.
| | - Bryan Mckiver
- Department of Pharmacology and Toxicology, and Translational Research Initiative for Pain and Neuropathy, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA.
| | - Wisam Toma
- Department of Pharmacology and Toxicology, and Translational Research Initiative for Pain and Neuropathy, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA.
| | - Andy Z X Zhu
- Takeda Pharmaceutical Company Limited, Cambridge, MA 02139, USA.
| | - Rachel F Tyndale
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Department of Pharmacology and Toxicology, and Psychiatry, University of Toronto, Toronto, ON, Canada.
| | - Vivek Kumar
- The Jackson Laboratory, Bar Harbor, ME 04609, USA.
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, and Translational Research Initiative for Pain and Neuropathy, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
42
|
Babbs RK, Beierle JA, Ruan QT, Kelliher JC, Chen MM, Feng AX, Kirkpatrick SL, Benitez FA, Rodriguez FA, Pierre JJ, Anandakumar J, Kumar V, Mulligan MK, Bryant CD. Cyfip1 Haploinsufficiency Increases Compulsive-Like Behavior and Modulates Palatable Food Intake in Mice: Dependence on Cyfip2 Genetic Background, Parent-of Origin, and Sex. G3 (BETHESDA, MD.) 2019; 9:3009-3022. [PMID: 31324746 PMCID: PMC6723122 DOI: 10.1534/g3.119.400470] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/18/2019] [Indexed: 12/11/2022]
Abstract
Binge eating (BE) is a heritable trait associated with eating disorders and involves episodes of rapid, large amounts of food consumption. We previously identified cytoplasmic FMR1-interacting protein 2 (Cyfip2) as a genetic factor underlying compulsive-like BE in mice. CYFIP2 is a homolog of CYFIP1 which is one of four paternally-deleted genes in patients with Type I Prader-Willi Syndrome (PWS), a neurodevelopmental disorder whereby 70% of cases involve paternal 15q11-q13 deletion. PWS symptoms include hyperphagia, obesity (if untreated), cognitive deficits, and obsessive-compulsive behaviors. We tested whether Cyfip1 haploinsufficiency (+/-) would enhance compulsive-like behavior and palatable food (PF) intake in a parental origin- and sex-dependent manner on two Cyfip2 genetic backgrounds, including the BE-prone C57BL/6N (Cyfip2N/N) background and the BE-resistant C57BL/6J (Cyfip2J/J) background. Cyfip1+/- mice showed increased compulsive-like behavior on both backgrounds and increased PF intake on the Cyfip2N/N background. In contrast, maternal Cyfip1 haploinsufficiency on the BE-resistant Cyfip2J/J background induced a robust escalation in PF intake in wild-type Cyfip1J/J males while having no effect in Cyfip1J/- males. Notably, induction of behavioral phenotypes in wild-type males following maternal Fmr1+/- has previously been reported. In the hypothalamus, there was a paternally-enhanced reduction in CYFIP1 protein whereas in the nucleus accumbens, there was a maternally-enhanced reduction in CYFIP1 protein. Nochange in FMR1 protein (FMRP) was observed in Cyfip1+/- mice, regardless of parental origin. To summarize, Cyfip1 haploinsufficiency increased compulsive-like behavior and induced genetic background-dependent, sex-dependent, and parent-of-origin-dependent effects on PF consumption and CYFIP1 expression that could have relevance for neurodevelopmental and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Richard K Babbs
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry
| | - Jacob A Beierle
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry
- T32 NIGMS Training Program in Biomolecular Pharmacology
- Boston University's Transformative Training Program in Addiction Science (TTPAS), Biomedical Genetics, Boston University School of Medicine, Boston, MA 02118
| | - Qiu T Ruan
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry
- T32 NIGMS Training Program in Biomolecular Pharmacology
- Boston University's Transformative Training Program in Addiction Science (TTPAS), Biomedical Genetics, Boston University School of Medicine, Boston, MA 02118
| | - Julia C Kelliher
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry
| | - Melanie M Chen
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry
| | - Ashley X Feng
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry
| | - Stacey L Kirkpatrick
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry
| | - Fabiola A Benitez
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry
| | - Fred A Rodriguez
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry
| | - Johanne J Pierre
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry
| | - Jeya Anandakumar
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry
| | - Vivek Kumar
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, and
| | - Megan K Mulligan
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, 71 S. Manassas St, Memphis, TN 38163
| | - Camron D Bryant
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry
| |
Collapse
|
43
|
Maloney SE, Rieger MA, Al-Hasani R, Bruchas MR, Wozniak DF, Dougherty JD. Loss of CELF6 RNA binding protein impairs cocaine conditioned place preference and contextual fear conditioning. GENES, BRAIN, AND BEHAVIOR 2019; 18:e12593. [PMID: 31215739 PMCID: PMC7059558 DOI: 10.1111/gbb.12593] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/13/2019] [Accepted: 06/02/2019] [Indexed: 12/21/2022]
Abstract
In addition to gene expression differences in distinct cell types, there is substantial post-transcriptional regulation driven in part by RNA binding proteins (RBPs). Loss-of-function RBP mutations have been associated with neurodevelopmental disorders, such as Fragile-X syndrome and syndromic autism. Work performed in animal models to elucidate the influence of neurodevelopmental disorder-associated RBPs on distinct behaviors has showed a connection between normal post-transcriptional regulation and conditioned learning. We previously reported cognitive inflexibility in a mouse model null for the RBP CUG-BP, Elav-like factor 6 (CELF6), which we also found to be associated with human autism. Specifically, these mice failed to potentiate exploratory hole-poking behavior in response to familiarization to a rewarding stimuli. Characterization of Celf6 gene expression showed high levels in monoaminergic populations such as the dopaminergic midbrain populations. To better understand the underlying behavioral disruption mediating the resistance to change exploratory behavior in the holeboard task, we tested three hypotheses: Does Celf6 loss lead to global restricted patterns of behavior, failure of immediate response to reward or failure to alter behavior in response to reward (conditioning). We found the acute response to reward was intact, yet Celf6 mutant mice exhibited impaired conditioned learning to both reward and aversive stimuli. Thus, we found that the resistance to change by the Celf6 mutant in the holeboard was most parsimoniously explained as a failure of conditioning, as the mice had blunted responses even to potent rewarding stimuli such as cocaine. These findings further support the role of RBPs in conditioned learning.
Collapse
Affiliation(s)
- Susan E. Maloney
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael A. Rieger
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ream Al-Hasani
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO 63110, USA
| | - Michael R. Bruchas
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David F. Wozniak
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joseph D. Dougherty
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
44
|
Terenina EE, Cavigelli S, Mormede P, Zhao W, Parks C, Lu L, Jones BC, Mulligan MK. Genetic Factors Mediate the Impact of Chronic Stress and Subsequent Response to Novel Acute Stress. Front Neurosci 2019; 13:438. [PMID: 31164799 PMCID: PMC6536627 DOI: 10.3389/fnins.2019.00438] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/16/2019] [Indexed: 12/31/2022] Open
Abstract
Individual differences in physiological and biobehavioral adaptation to chronic stress are important predictors of health and fitness; genetic differences play an important role in this adaptation. To identify these differences we measured the biometric, neuroendocrine, and transcriptional response to stress among inbred mouse strains with varying degrees of genetic similarity, C57BL/6J (B), C57BL/6NJ (N), and DBA/2J (D). The B and D strains are highly genetically diverse whereas the B and N substrains are highly similar. Strain differences in hypothalamic-pituitary-adrenal (HPA) axis cross-sensitization were determined by plasma corticosterone (CORT) levels and hippocampal gene expression following 7-weeks of chronic mild stress (CMS) or normal housing (NH) and subsequent exposure to novel acute restraint. Fecal CORT metabolites and body and organ weights were also measured. All strains exposed to CMS had reduced heart weights, whereas body weight gain was attenuated only in B and N strains. Acute stress alone produced larger plasma CORT responses in the D and N strains compared to the B strain. CMS paired with acute stress produced cross-sensitization of the CORT response in the N strain. The N strain also had the largest number of hippocampal transcripts with up-regulated expression in response to stress. In contrast, the D strain had the largest number of transcripts with down-regulated expression following CMS and acute stress. In summary, we observed differential responses to CMS at both the physiological and molecular level among genetically diverse strains, indicating that genetic factors drive individual differences in experience-dependent regulation of the stress response.
Collapse
Affiliation(s)
- Elena E Terenina
- GenPhySE, ENVT, INRA, Université de Toulouse, Castanet-Tolosan, France.,Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Sonia Cavigelli
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, United States
| | - Pierre Mormede
- GenPhySE, ENVT, INRA, Université de Toulouse, Castanet-Tolosan, France.,Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Wenyuan Zhao
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Cory Parks
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Byron C Jones
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Megan K Mulligan
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
45
|
Åhlgren J, Voikar V. Experiments done in Black-6 mice: what does it mean? Lab Anim (NY) 2019; 48:171-180. [DOI: 10.1038/s41684-019-0288-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 03/19/2019] [Indexed: 02/06/2023]
|
46
|
Zhao L, Mulligan MK, Nowak TS. Substrain- and sex-dependent differences in stroke vulnerability in C57BL/6 mice. J Cereb Blood Flow Metab 2019; 39:426-438. [PMID: 29260927 PMCID: PMC6421252 DOI: 10.1177/0271678x17746174] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The C57BL/6 mouse strain is represented by distinct substrains, increasingly recognized to differ genetically and phenotypically. The current study compared stroke vulnerability among C57BL/6 J (J), C57BL/6JEiJ (JEiJ), C57BL/6ByJ (ByJ), C57BL/6NCrl (NCrl), C57BL/6NJ (NJ) and C57BL/6NTac (NTac) substrains, using a model of permanent distal middle cerebral artery and common carotid artery occlusion. Mean infarct volume was nearly two-fold smaller in J, JEiJ and ByJ substrains relative to NCrl, NJ and NTac (N-lineage) mice. This identifies a previously unrecognized confound in stroke studies involving genetically modified strain comparisons if control substrain background were not rigorously matched. Mean infarct size was smaller in females of J and ByJ substrains than in the corresponding males, but there was no sex difference for NCrl and NJ mice. A higher proportion of small infarcts in J and ByJ substrains was largely responsible for both substrain- and sex-dependent differences. These could not be straightforwardly explained by variations in posterior communicating artery patency, MCA anatomy or acute penumbral blood flow deficits. Their larger and more homogeneously distributed infarcts, together with their established use as the common background for many genetically modified strains, may make N-lineage C57BL/6 substrains the preferred choice for future studies in experimental stroke.
Collapse
Affiliation(s)
- Liang Zhao
- 1 Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Megan K Mulligan
- 2 Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Thaddeus S Nowak
- 1 Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
47
|
Eisfeld AJ, Gasper DJ, Suresh M, Kawaoka Y. C57BL/6J and C57BL/6NJ Mice Are Differentially Susceptible to Inflammation-Associated Disease Caused by Influenza A Virus. Front Microbiol 2019; 9:3307. [PMID: 30713529 PMCID: PMC6346684 DOI: 10.3389/fmicb.2018.03307] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/19/2018] [Indexed: 01/01/2023] Open
Abstract
Influenza viruses cause seasonal epidemics and sporadic pandemics, and are a major burden on human health. To develop better countermeasures and improve influenza disease outcomes, a clearer understanding of influenza pathogenesis is necessary. Host genetic factors have emerged as potential regulators of human influenza disease susceptibility, and in the mouse model, genetic background has been clearly linked to influenza pathogenicity. Here, we show that C57BL/6J mice are significantly more susceptible to disease caused by a 2009 pandemic H1N1 virus, an H7N9 virus, and a highly pathogenic H5N1 influenza virus compared to the closely related substrain, C57BL/6NJ. Mechanistically, influenza virus infection in C57BL/6J mice results in earlier presentation of edema, increased immune cell infiltration, higher levels of inflammatory cytokines, greater tissue damage, and delayed activation of regenerative processes in infected lung tissues compared to C57BL/6NJ mice. These differences are not dependent on virus replication levels. Six genes with known coding region differences between C57BL/6J and C57BL/6NJ strains exhibit increased transcript levels in influenza virus-infected mouse lungs, suggesting potential contributions to regulation of disease susceptibility. This work uncovers a previously unappreciated difference in disease susceptibility between the closely related C57BL/6J and C57BL/6NJ mice, which may be exploited in future studies to identify host factors and/or specific genetic elements that regulate host-dependent inflammatory mechanisms involved in influenza virus pathogenicity.
Collapse
Affiliation(s)
- Amie J Eisfeld
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - David J Gasper
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - M Suresh
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States.,Division of Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
48
|
Bryant CD, Bagdas D, Goldberg LR, Khalefa T, Reed ER, Kirkpatrick SL, Kelliher JC, Chen MM, Johnson WE, Mulligan MK, Imad Damaj M. C57BL/6 substrain differences in inflammatory and neuropathic nociception and genetic mapping of a major quantitative trait locus underlying acute thermal nociception. Mol Pain 2019; 15:1744806918825046. [PMID: 30632432 PMCID: PMC6365993 DOI: 10.1177/1744806918825046] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 11/26/2018] [Accepted: 12/21/2018] [Indexed: 11/17/2022] Open
Abstract
Sensitivity to different pain modalities has a genetic basis that remains largely unknown. Employing closely related inbred mouse substrains can facilitate gene mapping of nociceptive behaviors in preclinical pain models. We previously reported enhanced sensitivity to acute thermal nociception in C57BL/6J (B6J) versus C57BL/6N (B6N) substrains. Here, we expanded on nociceptive phenotypes and observed an increase in formalin-induced inflammatory nociceptive behaviors and paw diameter in B6J versus B6N mice (Charles River Laboratories). No strain differences were observed in mechanical or thermal hypersensitivity or in edema following the Complete Freund's Adjuvant model of inflammatory pain, indicating specificity in the inflammatory nociceptive stimulus. In the chronic constrictive nerve injury, a model of neuropathic pain, no strain differences were observed in baseline mechanical threshold or in mechanical hypersensitivity up to one month post-chronic constrictive nerve injury. We replicated the enhanced thermal nociception in the 52.5°C hot plate test in B6J versus B6N mice from The Jackson Laboratory. Using a B6J × B6N-F2 cross (N = 164), we mapped a major quantitative trait locus underlying hot plate sensitivity to chromosome 7 that peaked at 26 Mb (log of the odds [LOD] = 3.81, p < 0.01; 8.74 Mb-36.50 Mb) that was more pronounced in males. Genes containing expression quantitative trait loci associated with the peak nociceptive marker that are implicated in pain and inflammation include Ryr1, Cyp2a5, Pou2f2, Clip3, Sirt2, Actn4, and Ltbp4 (false discovery rate < 0.05). Future studies involving positional cloning and gene editing will determine the quantitative trait gene(s) and potential pleiotropy of this locus across pain modalities.
Collapse
Affiliation(s)
- Camron D Bryant
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Deniz Bagdas
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
- Translational Research Initiative for Pain and Neuropathy, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
| | - Lisa R Goldberg
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
- Program in Biomolecular Pharmacology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Tala Khalefa
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
- Translational Research Initiative for Pain and Neuropathy, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
| | - Eric R Reed
- Department of Medicine, Computational Biomedicine, Bioinformatics Program, Boston University, Boston, MA, USA
| | - Stacey L Kirkpatrick
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Julia C Kelliher
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Melanie M Chen
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - William E Johnson
- Department of Medicine, Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Megan K Mulligan
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
- Translational Research Initiative for Pain and Neuropathy, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
49
|
Nowak TS, Mulligan MK. Impact of C57BL/6 substrain on sex-dependent differences in mouse stroke models. Neurochem Int 2018; 127:12-21. [PMID: 30448566 DOI: 10.1016/j.neuint.2018.11.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/14/2018] [Accepted: 11/14/2018] [Indexed: 01/18/2023]
Abstract
We have recently found significant variation in stroke vulnerability among substrains of C57BL/6 mice, observing that commonly used N-lineage substrains exhibit larger infarcts than C57BL/6J and related substrains. Parallel variation was also seen with respect to sex differences in stroke vulnerability, in that C57BL/6 mice of the N-lineage exhibited comparable infarct sizes in males and females, whereas infarcts tended to be smaller in females than in males of J-lineage substrains. This adds to the growing list of recognized phenotypic and genetic differences among C57BL/6 substrains. Although no previous studies have explicitly compared substrains with respect to sex differences in stroke vulnerability, unrecognized background mismatch has occurred in some studies involving control and genetically modified mice. The aims of this review are to: present the evidence for associated substrain- and sex-dependent differences in a mouse permanent occlusion stroke model; examine the extent to which the published literature in other models compares with these recent results; and consider the potential impact of unrecognized heterogeneity in substrain background on the interpretation of studies investigating the impact of genetic modifications on sex differences in stroke outcome. Substrain emerges as a critical variable to be documented in any experimental stroke study in mice.
Collapse
Affiliation(s)
- Thaddeus S Nowak
- Department of Neurology and Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA.
| | - Megan K Mulligan
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
50
|
Babbs RK, Kelliher JC, Scotellaro JL, Luttik KP, Mulligan MK, Bryant CD. Genetic differences in the behavioral organization of binge eating, conditioned food reward, and compulsive-like eating in C57BL/6J and DBA/2J strains. Physiol Behav 2018; 197:51-66. [PMID: 30261172 DOI: 10.1016/j.physbeh.2018.09.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 12/25/2022]
Abstract
Binge eating (BE) is a heritable symptom of eating disorders associated with anxiety, depression, malnutrition, and obesity. Genetic analysis of BE could facilitate therapeutic discovery. We used an intermittent, limited access BE paradigm involving sweetened palatable food (PF) to examine genetic differences in BE, conditioned food reward, and compulsive-like eating between C57BL/6J (B6J) and DBA/2J (D2J) inbred mouse strains. D2J mice showed a robust escalation in intake and conditioned place preference for the PF-paired side. D2J mice also showed a unique style of compulsive-like eating in the light/dark conflict test where they rapidly hoarded and consumed PF in the preferred unlit environment. BE and compulsive-like eating exhibited narrow-sense heritability estimates between 56 and 73%. To gain insight into the genetic basis, we phenotyped and genotyped a small cohort of 133 B6J × D2J-F2 mice at the peak location of three quantitative trait loci (QTL) previously identified in F2 mice for sweet taste (chromosome 4: 156 Mb), bitter taste (chromosome 6: 133 Mb) and behavioral sensitivity to drugs of abuse (chromosome 11: 50 Mb). The D2J allele on chromosome 6 was associated with greater PF intake on training days and greater compulsive-like PF intake, but only in males, suggesting that decreased bitter taste may increase BE in males. The D2J allele on chromosome 11 was associated with an increase in final PF intake and slope of escalation across days. Future studies employing larger crosses and genetic reference panels comprising B6J and D2J alleles will identify causal genes and neurobiological mechanisms.
Collapse
Affiliation(s)
- Richard K Babbs
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, United States
| | - Julia C Kelliher
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, United States
| | - Julia L Scotellaro
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, United States; Boston University Undergraduate Research Opportunity Program (UROP), United States
| | - Kimberly P Luttik
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, United States; Boston University Undergraduate Research Opportunity Program (UROP), United States
| | - Megan K Mulligan
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Camron D Bryant
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, United States.
| |
Collapse
|