4.7 Article

The Neurotrophic Factor Receptor p75 in the Rat Dorsolateral Striatum Drives Excessive Alcohol Drinking

Journal

JOURNAL OF NEUROSCIENCE
Volume 36, Issue 39, Pages 10116-10127

Publisher

SOC NEUROSCIENCE
DOI: 10.1523/JNEUROSCI.4597-14.2016

Keywords

addiction; alcohol; BDNF; dorsal striatum; neurotrophic factor; p75NTR

Categories

Funding

  1. NIAAA [R01 AA016848, R37 AA01684, P50 AA017072]
  2. Jean Perkins Foundation
  3. VA Merit Grant [I01BX000267]

Ask authors/readers for more resources

Brain-derived neurotrophic factor (BDNF) signaling in the dorsolateral striatum (DLS) keeps alcohol intake in moderation. For example, activation of the BDNF receptor tropomyosin receptor kinase B (TrkB) in the DLS reduces intake in rats that consume moderate amounts of alcohol. Here, we tested whether long-term excessive consumption of alcohol produces neuroadaptations in BDNF signaling in the rat DLS. We found that BDNF was no longer able to gate alcohol self-administration after a history of repeated cycles of binge alcohol drinking and withdrawal. We then elucidated the possible neuroadaptations that could block the ability of BDNF to keep consumption of alcohol in moderation. We report that intermittent access to 20% alcohol in a two-bottle choice paradigm that models excessive alcohol drinking produces a mobilization of DLS p75 neurotrophin receptor (p75NTR), whose activities oppose those of the Trk receptors, including TrkB. These neuroadaptations were not observed in the DLS of rats exposed to continuous access to 10% alcohol or in rats consuming sucrose. Furthermore, short hairpin RNA (shRNA)-mediated knockdown of the p75NTR gene in the DLS, as well as intra-DLS infusion or systemic administration of the p75NTR modulator, LM11A-31, significantly reduced binge drinking of alcohol. Together, our results suggest that excessive alcohol consumption produces a change in BDNF signaling in the DLS, which is mediated by the recruitment of p75NTR. Our data also imply that modulators of p75NTR signaling could be developed as medications for alcohol abuse disorders.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

Article Neurosciences

Ackr3-Venus knock-in mouse lights up brain vasculature

Aliza T. Ehrlich, Meriem Semache, Pierre Couvineau, Stefan Wojcik, Hiroyuki Kobayashi, Marcus Thelen, Florence Gross, Mireille Hogue, Christian Le Gouill, Emmanuel Darcq, Michel Bouvier, Brigitte L. Kieffer

Summary: Researchers have generated Ackr3-Venus knock-in mice to track the ACKR3 receptor, enabling direct detection of the receptor under physiological conditions and revealing its expression patterns in the brain.

MOLECULAR BRAIN (2021)

Article Biochemistry & Molecular Biology

Plasma tau, NfL, GFAP and UCHL1 as candidate biomarkers of alcohol withdrawal-associated brain damage: A pilot study

Virgile Clergue-Duval, Agathe Vrillon, Jerome Jeanblanc, Frank Questel, Julien Azuar, Gregory Fouquet, Francois Mouton-Liger, Dorian Rollet, Eric Hispard, Elodie Bouaziz-Amar, Vanessa Bloch, Alexandra Dereux, Emmanuel Cognat, Cynthia Marie-Claire, Jean-Louis Laplanche, Frank Bellivier, Claire Paquet, Mickael Naassila, Florence Vorspan

Summary: This translational study investigated the potential of plasma tau protein, neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP) and ubiquitin carboxy-terminal hydrolase L1 (UCHL1) as predictive biomarkers of alcohol withdrawal-associated brain toxicity. The study found that NfL levels were higher in the alcohol cessation group and tau and UCHL1 levels were positively associated with diazepam dosage. In a preclinical study, NfL levels were higher in the alcohol deprivation effect (ADE) model. Plasma tau, NfL and UCHL1 show promise as biomarkers of brain suffering during alcohol withdrawal.

ADDICTION BIOLOGY (2022)

Article Biochemistry & Molecular Biology

The GPR88 agonist RTI-13951-33 reduces alcohol drinking and seeking in mice

Sami Ben Hamida, Michelle Carter, Emmanuel Darcq, Marion Sourty, Md Toufiqur Rahman, Ann M. Decker, Chunyang Jin, Brigitte L. Kieffer

Summary: The study demonstrates that RTI-13951-33 can reduce alcohol drinking behavior in mice while leaving water drinking behavior intact. The compound is effective in C57BL/6 mice but not Gpr88 knockout mice, indicating its specificity in vivo. Additionally, RTI-13951-33 decreases alcohol self-administration and reduces the expression of alcohol reward-seeking behavior, suggesting its potential therapeutic effect for alcohol use disorders.

ADDICTION BIOLOGY (2022)

Article Neurosciences

Habenular Neurons Expressing Mu Opioid Receptors Promote Negative Affect in a Projection-Specific Manner

Julie Bailly, Florence Allain, Eric Schwartz, Chloe Tirel, Charles Dupuy, Florence Petit, Marco A. Diana, Emmanuel Darcq, Brigitte L. Kieffer

Summary: This study found that the mu opioid receptor in the brain can promote aversive emotional states, and it is controlled by two different neural pathways (Hb-MOR/interpeduncular nucleus and Hb-MOR/dorsal raphe nucleus) that regulate despair and anxiety, respectively. These findings are important for understanding emotional balance and addiction mechanisms.

BIOLOGICAL PSYCHIATRY (2023)

Article Endocrinology & Metabolism

Supraspinal melatonin MT2 receptor agonism alleviates pain via a neural circuit that recruits mu opioid receptors

Luca Posa, Danilo De Gregorio, Martha Lopez-Canul, Qianzi He, Emmanuel Darcq, Laura Rullo, Leora Pearl-Dowler, Livio Luongo, Sanzio Candeletti, Patrizia Romualdi, Brigitte Lina Kieffer, Gabriella Gobbi

Summary: In this study, the relationship between melatonin MT2 receptors and the opioid system in neuropathic pain was investigated. The results showed that the antiallodynic effects of a melatonin MT2 agonist required the activation of mu opioid receptors (MORs), but not delta opioid receptors (DORs). Electrophysiological recordings and immunohistochemistry studies revealed that the interaction between MORs and MT2 receptors within a neuronal circuit in the brain was responsible for the analgesic effects of the MT2 agonist.

JOURNAL OF PINEAL RESEARCH (2022)

Article Biochemistry & Molecular Biology

The orphan receptor GPR88 controls impulsivity and is a risk factor for Attention-Deficit/Hyperactivity Disorder

Sami Ben Hamida, Sarojini M. Sengupta, Ellie Clarke, Michael McNicholas, Eleonora Moroncini, Emmanuel Darcq, Marina Ter-Stepanian, Marie-Eve Fortier, Natalie Grizenko, Ridha Joober, Brigitte L. Kieffer

Summary: Both animal and human studies suggest that GPR88 is involved in Attention-Deficit/Hyperactivity Disorder (ADHD). Studies using Gpr88 knockout mice revealed ADHD-like behaviors, including increased motor impulsivity, reduced attention, and hyperactivity. The ADHD drug Atomoxetine reduced impulsivity in these mutant mice. Human genetic studies found associations between GPR88 variants and ADHD diagnosis, treatment response, and cognition, particularly in the presence of prenatal stress. Overall, GPR88 signaling appears to be a key factor in the diagnosis and treatment of ADHD.

MOLECULAR PSYCHIATRY (2022)

Article Chemistry, Medicinal

Synthesis and pharmacological validation of a novel radioligand for the orphan GPR88 receptor

Ann M. Decker, Md Toufiqur Rahman, Chad M. Kormos, David Hesk, Emmanuel Darcq, Brigitte L. Kieffer, Chunyang Jin

Summary: This study synthesized and characterized the first GPR88 radio-ligand, [3H]RTI-33, derived from a synthetic agonist RTI-13951-33. [3H]RTI-33 showed specific and saturable binding (KD of 85 nM) in membranes prepared from stable PPLS-HA-hGPR88-CHO cells, and a competition binding assay was developed to determine binding affinities of several known GPR88 agonists. This radioligand represents a powerful tool for future mechanistic and cell-based ligand-receptor interaction studies of GPR88.

BIOORGANIC & MEDICINAL CHEMISTRY LETTERS (2023)

Article Neurosciences

Reduced dopamine release in Dcc haploinsufficiency male mice abolishes the rewarding effects of cocaine but not those of morphine and ethanol

Emmanuel Darcq, Dominique Nouel, Giovanni Hernandez, Matthew Pokinko, Polina Ash, Luc Moquin, Alain Gratton, Brigitte Kieffer, Cecilia Flores

Summary: The protective effects of Dcc haploinsufficiency against the rewarding effects of stimulant drugs do not extend to opioids and ethanol, suggesting different brain circuits mediate the rewarding effects of these substances.

PSYCHOPHARMACOLOGY (2023)

Article Multidisciplinary Sciences

The mu opioid receptor and the orphan receptor GPR151 contribute to social reward in the habenula

Florence Allain, Michelle Carter, Sylvie Dumas, Emmanuel Darcq, Brigitte L. Kieffer

Summary: The mu opioid receptor (MOR) and the orphan GPR151 receptor in the habenula are found to play important roles in social behaviors, and their absence leads to impaired social interactions in mice.

SCIENTIFIC REPORTS (2022)

Article Neurosciences

Mu Opioid Receptor-Expressing Neurons in the Dorsal Raphe Nucleus Are Involved in Reward Processing and Affective Behaviors

Lola Welsch, Esther Colantonio, Mathilde Frison, Desiree A. Johnson, Shannan P. Mcclain, Victor Mathis, Matthew R. Banghart, Sami Ben Hamida, Emmanuel Darcq, Brigitte L. Kieffer

Summary: This study found that μ opioid receptor neurons in the dorsal raphe nucleus (DRN) participate in reward and emotional responses. These neurons respond to rewarding stimuli and their optoactivation has reinforcing effects and promotes positive emotional responses. Furthermore, their effects are partially mediated by their projections to the lateral hypothalamus.

BIOLOGICAL PSYCHIATRY (2023)

Article Chemistry, Medicinal

Improvement of the Metabolic Stability of GPR88 Agonist RTI-13951-33: Design, Synthesis, and Biological Evaluation

Md Toufiqur Rahman, Ann M. Decker, Sami Ben Hamida, David A. Perrey, Hetti Handi Chaminda Lakmal, Rangan Maitra, Emmanuel Darcq, Brigitte L. Kieffer, Chunyang Jin

Summary: We reported a new GPR88 agonist, RTI-122, with high potency and good metabolic stability, which could improve alcohol drinking behavior. This suggests that RTI-122 may serve as a promising lead compound for GPR88 agonist drug discovery research.

JOURNAL OF MEDICINAL CHEMISTRY (2023)

Article Cell Biology

Striatal m-opioid receptor activation triggers direct-pathway GABAergic plasticity and induces negative affect

Wei Wang, Xueyi Xie, Xiaowen Zhuang, Yufei Huang, Tao Tan, Himanshu Gangal, Zhenbo Huang, William Purvines, Xuehua Wang, Alexander Stefanov, Ruifeng Chen, Lucas Rodriggs, Anita Chaiprasert, Emily Yu, Valerie Vierkant, Michelle Hook, Yun Huang, Emmanuel Darcq, Jun Wang

Summary: Withdrawal from chronic opioid use leads to hypodopaminergic states and negative affect, promoting relapse. Activation of MORs in dMSNs in the striatal patch compartment suppresses striatopallidal transmission, and withdrawal potentiated this transmission. Fentanyl self-administration enhances striatonigral transmission and reduces dopaminergic activity, while activated striatal neurons mediate contextual memory retrieval. Inhibition of striatal MOR+ neurons rescues fentanyl withdrawal-induced physical symptoms and anxiety-like behaviors. These findings suggest that chronic opioid use induces GABAergic striatopallidal and striatonigral plasticity, leading to hypodopaminergic states and relapse.

CELL REPORTS (2023)

Article Psychiatry

Chronic tianeptine induces tolerance in analgesia and hyperlocomotion via mu-opioid receptor activation in mice

Florence Allain, Aliza T. Ehrlich, Michael McNicholas, Florence Gross, Weiya Ma, Brigitte L. Kieffer, Emmanuel Darcq

Summary: This study found that the antidepressant tianeptine mainly exerts its effects on the brain through the μ-opioid receptor, and it exhibits analgesic, locomotor, and rewarding behaviors in animal models. These effects can only be observed in mice with positive expression of the opioid receptor, and are ineffective in mice with negative expression. In addition, chronic use of tianeptine may lead to tolerance to its analgesic and hyperlocomotor effects.

FRONTIERS IN PSYCHIATRY (2023)

Article Neurosciences

Advances in the characterization of negative affect caused by acute and protracted opioid withdrawal using animal models

Dersu Ozdemir, Florence Allain, Brigitte L. Kieffer, Emmanuel Darcq

Summary: Opioid use disorder is a chronic brain disease caused by long-term neuroadaptations resulting from repeated opioid consumption and withdrawal. These neuroadaptations lead to negative affect and various symptoms, such as loss of motivation, anxiety, social deficits, heightened stress reactivity, emotional and physical pain, dysphoria, sleep disorders, and chronic irritability. Understanding the neurocircuitry involved in withdrawal and abstinence is crucial for treatment and relapse prevention.

NEUROPHARMACOLOGY (2023)

Article Neurosciences

Mu Opioid Receptor-Positive Neurons in the Dorsal Raphe Nucleus Are Impaired by Morphine Abstinence

Lola Welsch, Esther Colantonio, Camille Falconnier, Cedric Champagnol-DiLiberti, Florence Allain, Sami Ben Hamida, Emmanuel Darcq, Pierre-Eric Lutz, Brigitte L. Kieffer

Summary: Chronic morphine abstinence leads to reduced MOR function in DRN-MOR neurons and abnormal self-stimulation, which may increase propensity for addiction-related behaviors.

BIOLOGICAL PSYCHIATRY (2023)

No Data Available