Publications from the Electron Microscopy Core. Zhu, Xingliang; Joo, Yuyoung; Bossi, Simone; McDevitt, Ross A; Xie, Aoji; Wang, Yue; Xue, Yutong; Su, Shuaikun; Lee, Seung Kyu; Sah, Nirnath; Zhang, Shiliang; Ye, Rong; Pinto, Alejandro; Zhang, Yongqing; Araki, Kimi; Araki, Masatake; Morales, Marisela; Mattson, Mark P; van Praag, Henriette; Wang, Weidong Tdrd3-null mice show post-transcriptional and behavioral impairments associated with neurogenesis and synaptic plasticity Journal Article In: Prog Neurobiol, vol. 233, pp. 102568, 2024, ISSN: 1873-5118. Barbano, M Flavia; Zhang, Shiliang; Chen, Emma; Espinoza, Orlando; Mohammad, Uzma; Alvarez-Bagnarol, Yocasta; Liu, Bing; Hahn, Suyun; Morales, Marisela Lateral hypothalamic glutamatergic inputs to VTA glutamatergic neurons mediate prioritization of innate defensive behavior over feeding Journal Article In: Nat Commun, vol. 15, no. 1, pp. 403, 2024, ISSN: 2041-1723. Barker, David J; Zhang, Shiliang; Wang, Huiling; Estrin, David J; Miranda-Barrientos, Jorge; Liu, Bing; Kulkarni, Rucha J; de Deus, Junia Lara; Morales, Marisela Lateral preoptic area glutamate neurons relay nociceptive information to the ventral tegmental area Journal Article In: Cell Rep, vol. 42, no. 9, pp. 113029, 2023, ISSN: 2211-1247. Salinas, Armando G; Lee, Jeong Oen; Augustin, Shana M; Zhang, Shiliang; Patriarchi, Tommaso; Tian, Lin; Morales, Marisela; Mateo, Yolanda; Lovinger, David M Distinct sub-second dopamine signaling in dorsolateral striatum measured by a genetically-encoded fluorescent sensor Journal Article In: Nat Commun, vol. 14, no. 1, pp. 5915, 2023, ISSN: 2041-1723. Bonaventura, Jordi; Boehm, Matthew A; Jedema, Hank P; Solis, Oscar; Pignatelli, Marco; Song, Xiaowei; Lu, Hanbing; Richie, Christopher T; Zhang, Shiliang; Gomez, Juan L; Lam, Sherry; Morales, Marisela; Gharbawie, Omar A; Pomper, Martin G; Stein, Elliot A; Bradberry, Charles W; Michaelides, Michael Expression of the excitatory opsin ChRERα can be traced longitudinally in rat and nonhuman primate brains with PET imaging Journal Article In: Sci Transl Med, vol. 15, no. 706, pp. eadd1014, 2023, ISSN: 1946-6242. Zhang, Shiliang Steven; Shevelkin, Alexey; Yu, Kevin; Wang, Huiling; Morales, Marisela In: Microsc Microanal, vol. 29, no. 29 Suppl 1, pp. 1192–1194, 2023, ISSN: 1435-8115. Rubio, F Javier; Olivares, Daniel E; Dunn, Christopher; Zhang, Shiliang; Hilaire, Elias M; Henry, Akeem; Mejias-Aponte, Carlos; Nogueras-Ortiz, Carlos J; Selvam, Pooja V; Cruz, Fabio C; Madangopal, Rajtarun; Morales, Marisela; Hope, Bruce T In: J Neurosci, vol. 43, no. 23, pp. 4217–4233, 2023, ISSN: 1529-2401. Solis, Oscar; Beccari, Andrea R; Iaconis, Daniela; Talarico, Carmine; Ruiz-Bedoya, Camilo A; Nwachukwu, Jerome C; Cimini, Annamaria; Castelli, Vanessa; Bertini, Riccardo; Montopoli, Monica; Cocetta, Veronica; Borocci, Stefano; Prandi, Ingrid G; Flavahan, Kelly; Bahr, Melissa; Napiorkowski, Anna; Chillemi, Giovanni; Ooka, Masato; Yang, Xiaoping; Zhang, Shiliang; Xia, Menghang; Zheng, Wei; Bonaventura, Jordi; Pomper, Martin G; Hooper, Jody E; Morales, Marisela; Rosenberg, Avi Z; Nettles, Kendall W; Jain, Sanjay K; Allegretti, Marcello; Michaelides, Michael The SARS-CoV-2 spike protein binds and modulates estrogen receptors Journal Article In: Sci Adv, vol. 8, no. 48, pp. eadd4150, 2022, ISSN: 2375-2548. Miranda-Barrientos, Jorge; Chambers, Ian; Mongia, Smriti; Liu, Bing; Wang, Hui-Ling; Mateo-Semidey, Gabriel E; Margolis, Elyssa B; Zhang, Shiliang; Morales, Marisela In: European Journal of Neuroscience, vol. 54, no. 1, pp. 4061-4084, 2021. Yang, Beimeng; Dan, Xiuli; Hou, Yujun; Lee, Jong-Hyuk; Wechter, Noah; Krishnamurthy, Sudarshan; Kimura, Risako; Babbar, Mansi; Demarest, Tyler; McDevitt, Ross; Zhang, Shiliang; Zhang, Yongqing; Mattson, Mark P; Croteau, Deborah L; Bohr, Vilhelm A NAD+ supplementation prevents STING-induced senescence in ataxia telangiectasia by improving mitophagy Journal Article In: Aging Cell, vol. 20, no. 4, pp. e13329, 2021, ISSN: 1474-9726. Barbano, Flavia M; Wang, Hui-Ling; Zhang, Shiliang; Miranda-Barrientos, Jorge; Estrin, David J; Figueroa-González, Almaris; Liu, Bing; Barker, David J; Morales, Marisela VTA Glutamatergic Neurons Mediate Innate Defensive Behaviors Journal Article In: Neuron, vol. 107, no. 2, pp. 368–382.e8, 2020, ISBN: 0896-6273. Root, David H; Barker, David J; Estrin, David J; Miranda-Barrientos, Jorge A; Liu, Bing; Zhang, Shiliang; Wang, Hui-Ling; Vautier, Francois; Ramakrishnan, Charu; Kim, Yoon Seok; Fenno, Lief; Deisseroth, Karl; Morales, Marisela Distinct Signaling by Ventral Tegmental Area Glutamate, GABA, and Combinatorial Glutamate-GABA Neurons in Motivated Behavior Journal Article In: Cell Rep, vol. 32, no. 9, pp. 108094, 2020, ISSN: 2211-1247. Mongia, Smriti; Yamaguchi, Tsuyoshi; Liu, Bing; Zhang, Shiliang; Wang, Huiling; Morales, Marisela The Ventral Tegmental Area has calbindin neurons with the capability to co-release glutamate and dopamine into the nucleus accumbens. Journal Article In: Eur J Neurosci, 2019, ISSN: 1460-9568 (Electronic); 0953-816X (Linking). Zhang, Shiliang; Morales, Marisela Ultrastructural Detection of Neuronal Markers, Receptors, and Vesicular Transporters Journal Article In: Current Protocols in Neuroscience, vol. 88, no. 1, 2019. Zhang, Peisu; Kishimoto, Yuki; Grammatikakis, Ioannis; Gottimukkala, Kamalvishnu; Cutler, Roy G; Zhang, Shiliang; Abdelmohsen, Kotb; Bohr, Vilhelm A; Sen, Jyoti Misra; Gorospe, Myriam; Mattson, Mark P Senolytic therapy alleviates Abeta-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer's disease model. Journal Article In: Nat Neurosci, vol. 22, no. 5, pp. 719–728, 2019, ISSN: 1546-1726 (Electronic); 1097-6256 (Linking). Wang, Hui-Ling; Zhang, Shiliang; Qi, Jia; Wang, Huikun; Cachope, Roger; Mejias-Aponte, Carlos A; Gomez, Jorge A; Mateo-Semidey, Gabriel E; Beaudoin, Gerard M J; Paladini, Carlos A; Cheer, Joseph F; Morales, Marisela Dorsal Raphe Dual Serotonin-Glutamate Neurons Drive Reward by Establishing Excitatory Synapses on VTA Mesoaccumbens Dopamine Neurons. Journal Article In: Cell Rep, vol. 26, no. 5, pp. 1128–1142, 2019, ISSN: 2211-1247 (Electronic). Root, David H; Zhang, Shiliang; Barker, David J; Miranda-Barrientos, Jorge; Liu, Bing; Wang, Hui-Ling; Morales, Marisela Selective Brain Distribution and Distinctive Synaptic Architecture of Dual Glutamatergic-GABAergic Neurons. Journal Article In: Cell Rep, vol. 23, no. 12, pp. 3465–3479, 2018, ISSN: 2211-1247 (Electronic). Mateo, Yolanda; Johnson, Kari A; Covey, Dan P; Atwood, Brady K; Wang, Hui-Ling; Zhang, Shiliang; Gildish, Iness; Cachope, Roger; Bellocchio, Luigi; Guzman, Manuel; Morales, Marisela; Cheer, Joseph F; Lovinger, David M Endocannabinoid Actions on Cortical Terminals Orchestrate Local Modulation of Dopamine Release in the Nucleus Accumbens. Journal Article In: Neuron, vol. 96, no. 5, pp. 1112–1126, 2017, ISSN: 1097-4199 (Electronic); 0896-6273 (Linking). Barker, David J; Miranda-Barrientos, Jorge; Zhang, Shiliang; Root, David H; Wang, Hui-Ling; Liu, Bing; Calipari, Erin S; Morales, Marisela Lateral Preoptic Control of the Lateral Habenula through Convergent Glutamate and GABA Transmission. Journal Article In: Cell Rep, vol. 21, no. 7, pp. 1757–1769, 2017, ISSN: 2211-1247 (Electronic). Venniro, Marco; Caprioli, Daniele; Zhang, Michelle; Whitaker, Leslie R; Zhang, Shiliang; Warren, Brandon L; Cifani, Carlo; Marchant, Nathan J; Yizhar, Ofer; Bossert, Jennifer M; Chiamulera, Cristiano; Morales, Marisela; Shaham, Yavin The Anterior Insular Cortex-->Central Amygdala Glutamatergic Pathway Is Critical to Relapse after Contingency Management. Journal Article In: Neuron, vol. 96, no. 2, pp. 414–427, 2017, ISSN: 1097-4199 (Electronic); 0896-6273 (Linking). Edwards, Nicholas J; Tejeda, Hugo A; Pignatelli, Marco; Zhang, Shiliang; McDevitt, Ross A; Wu, Jocelyn; Bass, Caroline E; Bettler, Bernhard; Morales, Marisela; Bonci, Antonello Circuit specificity in the inhibitory architecture of the VTA regulates cocaine-induced behavior. Journal Article In: Nat Neurosci, vol. 20, no. 3, pp. 438–448, 2017, ISSN: 1546-1726 (Electronic); 1097-6256 (Linking). Steidl, Stephan; Wang, Huiling; Ordonez, Marco; Zhang, Shiliang; Morales, Marisela Optogenetic excitation in the ventral tegmental area of glutamatergic or cholinergic inputs from the laterodorsal tegmental area drives reward. Journal Article In: Eur J Neurosci, vol. 45, no. 4, pp. 559–571, 2017, ISSN: 1460-9568 (Electronic); 0953-816X (Linking).
2024
@article{pmid38216113,
title = {Tdrd3-null mice show post-transcriptional and behavioral impairments associated with neurogenesis and synaptic plasticity},
author = {Xingliang Zhu and Yuyoung Joo and Simone Bossi and Ross A McDevitt and Aoji Xie and Yue Wang and Yutong Xue and Shuaikun Su and Seung Kyu Lee and Nirnath Sah and Shiliang Zhang and Rong Ye and Alejandro Pinto and Yongqing Zhang and Kimi Araki and Masatake Araki and Marisela Morales and Mark P Mattson and Henriette van Praag and Weidong Wang},
url = {https://pubmed.ncbi.nlm.nih.gov/38216113/},
doi = {10.1016/j.pneurobio.2024.102568},
issn = {1873-5118},
year = {2024},
date = {2024-02-01},
urldate = {2024-02-01},
journal = {Prog Neurobiol},
volume = {233},
pages = {102568},
abstract = {The Topoisomerase 3B (Top3b) - Tudor domain containing 3 (Tdrd3) protein complex is the only dual-activity topoisomerase complex that can alter both DNA and RNA topology in animals. TOP3B mutations in humans are associated with schizophrenia, autism and cognitive disorders; and Top3b-null mice exhibit several phenotypes observed in animal models of psychiatric and cognitive disorders, including impaired cognitive and emotional behaviors, aberrant neurogenesis and synaptic plasticity, and transcriptional defects. Similarly, human TDRD3 genomic variants have been associated with schizophrenia, verbal short-term memory and educational attainment. However, the importance of Tdrd3 in normal brain function has not been examined in animal models. Here we generated a Tdrd3-null mouse strain and demonstrate that these mice display both shared and unique defects when compared to Top3b-null mice. Shared defects were observed in cognitive behaviors, synaptic plasticity, adult neurogenesis, newborn neuron morphology, and neuronal activity-dependent transcription; whereas defects unique to Tdrd3-deficient mice include hyperactivity, changes in anxiety-like behaviors, olfaction, increased new neuron complexity, and reduced myelination. Interestingly, multiple genes critical for neurodevelopment and cognitive function exhibit reduced levels in mature but not nascent transcripts. We infer that the entire Top3b-Tdrd3 complex is essential for normal brain function, and that defective post-transcriptional regulation could contribute to cognitive and psychiatric disorders.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{pmid38195566,
title = {Lateral hypothalamic glutamatergic inputs to VTA glutamatergic neurons mediate prioritization of innate defensive behavior over feeding},
author = {M Flavia Barbano and Shiliang Zhang and Emma Chen and Orlando Espinoza and Uzma Mohammad and Yocasta Alvarez-Bagnarol and Bing Liu and Suyun Hahn and Marisela Morales},
url = {https://pubmed.ncbi.nlm.nih.gov/38195566/},
doi = {10.1038/s41467-023-44633-w},
issn = {2041-1723},
year = {2024},
date = {2024-01-01},
urldate = {2024-01-01},
journal = {Nat Commun},
volume = {15},
number = {1},
pages = {403},
abstract = {The lateral hypothalamus (LH) is involved in feeding behavior and defense responses by interacting with different brain structures, including the Ventral Tegmental Area (VTA). Emerging evidence indicates that LH-glutamatergic neurons infrequently synapse on VTA-dopamine neurons but preferentially establish multiple synapses on VTA-glutamatergic neurons. Here, we demonstrated that LH-glutamatergic inputs to VTA promoted active avoidance, long-term aversion, and escape attempts. By testing feeding in the presence of a predator, we observed that ongoing feeding was decreased, and that this predator-induced decrease in feeding was abolished by photoinhibition of the LH-glutamatergic inputs to VTA. By VTA specific neuronal ablation, we established that predator-induced decreases in feeding were mediated by VTA-glutamatergic neurons but not by dopamine or GABA neurons. Thus, we provided evidence for an unanticipated neuronal circuitry between LH-glutamatergic inputs to VTA-glutamatergic neurons that plays a role in prioritizing escape, and in the switch from feeding to escape in mice.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
2023
@article{pmid37632750b,
title = {Lateral preoptic area glutamate neurons relay nociceptive information to the ventral tegmental area},
author = {David J Barker and Shiliang Zhang and Huiling Wang and David J Estrin and Jorge Miranda-Barrientos and Bing Liu and Rucha J Kulkarni and Junia Lara de Deus and Marisela Morales},
url = {https://pubmed.ncbi.nlm.nih.gov/37632750/},
doi = {10.1016/j.celrep.2023.113029},
issn = {2211-1247},
year = {2023},
date = {2023-09-01},
urldate = {2023-09-01},
journal = {Cell Rep},
volume = {42},
number = {9},
pages = {113029},
abstract = {The ventral tegmental area (VTA) has been proposed to play a role in pain, but the brain structures modulating VTA activity in response to nociceptive stimuli remain unclear. Here, we demonstrate that the lateral preoptic area (LPO) glutamate neurons relay nociceptive information to the VTA. These LPO glutamatergic neurons synapsing on VTA neurons respond to nociceptive stimulation and conditioned stimuli predicting nociceptive stimulation and also mediate aversion. In contrast, LPO GABA neurons synapsing in the VTA mediate reward. By ultrastructural quantitative synaptic analysis, ex vivo electrophysiology, and functional neuroanatomy we identify a complex circuitry between LPO glutamatergic and GABAergic neurons and VTA dopaminergic, GABAergic, and glutamatergic neurons. We conclude that LPO glutamatergic neurons play a causal role in the processing of nociceptive stimuli and in relaying information about nociceptive stimuli. The pathway from LPO glutamatergic neurons to the VTA represents an unpredicted interface between peripheral nociceptive information and the limbic system.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{pmid37739964,
title = {Distinct sub-second dopamine signaling in dorsolateral striatum measured by a genetically-encoded fluorescent sensor},
author = {Armando G Salinas and Jeong Oen Lee and Shana M Augustin and Shiliang Zhang and Tommaso Patriarchi and Lin Tian and Marisela Morales and Yolanda Mateo and David M Lovinger},
url = {https://pubmed.ncbi.nlm.nih.gov/37739964/},
doi = {10.1038/s41467-023-41581-3},
issn = {2041-1723},
year = {2023},
date = {2023-09-01},
urldate = {2023-09-01},
journal = {Nat Commun},
volume = {14},
number = {1},
pages = {5915},
abstract = {The development of genetically encoded dopamine sensors such as dLight has provided a new approach to measuring slow and fast dopamine dynamics both in brain slices and in vivo, possibly enabling dopamine measurements in areas like the dorsolateral striatum (DLS) where previously such recordings with fast-scan cyclic voltammetry (FSCV) were difficult. To test this, we first evaluated dLight photometry in mouse brain slices with simultaneous FSCV and found that both techniques yielded comparable results, but notable differences in responses to dopamine transporter inhibitors, including cocaine. We then used in vivo fiber photometry with dLight in mice to examine responses to cocaine in DLS. We also compared dopamine responses during Pavlovian conditioning across the striatum. We show that dopamine increases were readily detectable in DLS and describe transient dopamine kinetics, as well as slowly developing signals during conditioning. Overall, our findings indicate that dLight photometry is well suited to measuring dopamine dynamics in DLS.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{pmid37494470b,
title = {Expression of the excitatory opsin ChRERα can be traced longitudinally in rat and nonhuman primate brains with PET imaging},
author = {Jordi Bonaventura and Matthew A Boehm and Hank P Jedema and Oscar Solis and Marco Pignatelli and Xiaowei Song and Hanbing Lu and Christopher T Richie and Shiliang Zhang and Juan L Gomez and Sherry Lam and Marisela Morales and Omar A Gharbawie and Martin G Pomper and Elliot A Stein and Charles W Bradberry and Michael Michaelides},
url = {https://pubmed.ncbi.nlm.nih.gov/37494470/},
doi = {10.1126/scitranslmed.add1014},
issn = {1946-6242},
year = {2023},
date = {2023-07-01},
urldate = {2023-07-01},
journal = {Sci Transl Med},
volume = {15},
number = {706},
pages = {eadd1014},
abstract = {Optogenetics is a widely used technology with potential for translational research. A critical component of such applications is the ability to track the location of the transduced opsin in vivo. To address this problem, we engineered an excitatory opsin, ChRERα (hChR2(134R)-V5-ERα-LBD), that could be visualized using positron emission tomography (PET) imaging in a noninvasive, longitudinal, and quantitative manner. ChRERα consists of the prototypical excitatory opsin channelrhodopsin-2 (ChR2) and the ligand-binding domain (LBD) of the human estrogen receptor α (ERα). ChRERα showed conserved ChR2 functionality and high affinity for [F]16α-fluoroestradiol (FES), an FDA-approved PET radiopharmaceutical. Experiments in rats demonstrated that adeno-associated virus (AAV)-mediated expression of ChRERα enables neural circuit manipulation in vivo and that ChRERα expression could be monitored using FES-PET imaging. In vivo experiments in nonhuman primates (NHPs) confirmed that ChRERα expression could be monitored at the site of AAV injection in the primary motor cortex and in long-range neuronal terminals for up to 80 weeks. The anatomical connectivity map of the primary motor cortex identified by FES-PET imaging of ChRERα expression overlapped with a functional connectivity map identified using resting state fMRI in a separate cohort of NHPs. Overall, our results demonstrate that ChRERα expression can be mapped longitudinally in the mammalian brain using FES-PET imaging and can be used for neural circuit modulation in vivo.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{pmid37613243,
title = {Corelative Light and Electron Microscopic Study on 3D Reconstruction of Lateral Habenula Single Co-releasing GABA-Glutamate Axon Terminals Establishing Converging Synapses for Glutamate or GABA Release},
author = {Shiliang Steven Zhang and Alexey Shevelkin and Kevin Yu and Huiling Wang and Marisela Morales},
url = {https://pubmed.ncbi.nlm.nih.gov/37613243/},
doi = {10.1093/micmic/ozad067.614},
issn = {1435-8115},
year = {2023},
date = {2023-07-01},
urldate = {2023-07-01},
journal = {Microsc Microanal},
volume = {29},
number = {29 Suppl 1},
pages = {1192--1194},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{pmid37160369,
title = {Flow Cytometry of Synaptoneurosomes (FCS) Reveals Increased Ribosomal S6 and Calcineurin Proteins in Activated Medial Prefrontal Cortex to Nucleus Accumbens Synapses},
author = {F Javier Rubio and Daniel E Olivares and Christopher Dunn and Shiliang Zhang and Elias M Hilaire and Akeem Henry and Carlos Mejias-Aponte and Carlos J Nogueras-Ortiz and Pooja V Selvam and Fabio C Cruz and Rajtarun Madangopal and Marisela Morales and Bruce T Hope},
url = {https://pubmed.ncbi.nlm.nih.gov/37160369/},
doi = {10.1523/JNEUROSCI.0927-22.2023},
issn = {1529-2401},
year = {2023},
date = {2023-06-01},
urldate = {2023-06-01},
journal = {J Neurosci},
volume = {43},
number = {23},
pages = {4217--4233},
abstract = {Learning and behavior activate cue-specific patterns of sparsely distributed cells and synapses called ensembles that undergo memory-encoding engram alterations. While Fos is often used to label selectively activated cell bodies and identify neuronal ensembles, there is no comparable endogenous marker to label activated synapses and identify synaptic ensembles. For the purpose of identifying candidate synaptic activity markers, we optimized a flow cytometry of synaptoneurosome (FCS) procedure for assessing protein alterations in activated synapses from male and female rats. After injecting yellow fluorescent protein (YFP)-expressing adeno-associated virus into medial prefrontal cortex (mPFC) to label terminals in nucleus accumbens (NAc) of rats, we injected 20 mg/kg cocaine in a novel context (cocaine+novelty) to activate synapses, and prepared NAc synaptoneurosomes 0-60 min following injections. For FCS, we used commercially available antibodies to label presynaptic and postsynaptic markers synaptophysin and PSD-95 as well as candidate markers of synaptic activity [activity-regulated cytoskeleton protein (Arc), CaMKII and phospho-CaMKII, ribosomal protein S6 (S6) and phospho-S6, and calcineurin and phospho-calcineurin] in YFP-labeled synaptoneurosomes. Cocaine+novelty increased the percentage of S6-positive synaptoneurosomes at 5-60 min and calcineurin-positive synaptoneurosomes at 5-10 min. Electron microscopy verified that S6 and calcineurin levels in synaptoneurosomes were increased 10 min after cocaine+novelty. Pretreatment with the anesthetic chloral hydrate blocked cocaine+novelty-induced S6 and calcineurin increases in synaptoneurosomes, and novel context exposure alone (without cocaine) increased S6, both of which indicate that these increases were due to neural activity per se. Overall, FCS can be used to study protein alterations in activated synapses coming from specifically labeled mPFC projections to NAc. Memories are formed during learning and are stored in the brain by long-lasting molecular and cellular alterations called engrams formed within specific patterns of cue-activated neurons called neuronal ensembles. While Fos has been used to identify activated ensemble neurons and the engrams within them, we have not had a similar marker for activated synapses that can be used to identify synaptic engrams. Here we developed a procedure for high-throughput in-line analysis of flow cytometry of synaptoneurosome (FCS) and found that ribosomal S6 protein and calcineurin were increased in activated mPFC-NAc synapses. FCS can be used to study protein alterations in activated synapses within specifically labeled circuits.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
2022
@article{pmid36449624b,
title = {The SARS-CoV-2 spike protein binds and modulates estrogen receptors},
author = {Oscar Solis and Andrea R Beccari and Daniela Iaconis and Carmine Talarico and Camilo A Ruiz-Bedoya and Jerome C Nwachukwu and Annamaria Cimini and Vanessa Castelli and Riccardo Bertini and Monica Montopoli and Veronica Cocetta and Stefano Borocci and Ingrid G Prandi and Kelly Flavahan and Melissa Bahr and Anna Napiorkowski and Giovanni Chillemi and Masato Ooka and Xiaoping Yang and Shiliang Zhang and Menghang Xia and Wei Zheng and Jordi Bonaventura and Martin G Pomper and Jody E Hooper and Marisela Morales and Avi Z Rosenberg and Kendall W Nettles and Sanjay K Jain and Marcello Allegretti and Michael Michaelides},
url = {https://pubmed.ncbi.nlm.nih.gov/36449624/},
doi = {10.1126/sciadv.add4150},
issn = {2375-2548},
year = {2022},
date = {2022-12-01},
urldate = {2022-12-01},
journal = {Sci Adv},
volume = {8},
number = {48},
pages = {eadd4150},
abstract = {The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein binds angiotensin-converting enzyme 2 as its primary infection mechanism. Interactions between S and endogenous proteins occur after infection but are not well understood. We profiled binding of S against >9000 human proteins and found an interaction between S and human estrogen receptor α (ERα). Using bioinformatics, supercomputing, and experimental assays, we identified a highly conserved and functional nuclear receptor coregulator (NRC) LXD-like motif on the S2 subunit. In cultured cells, S DNA transfection increased ERα cytoplasmic accumulation, and S treatment induced ER-dependent biological effects. Non-invasive imaging in SARS-CoV-2-infected hamsters localized lung pathology with increased ERα lung levels. Postmortem lung experiments from infected hamsters and humans confirmed an increase in cytoplasmic ERα and its colocalization with S in alveolar macrophages. These findings describe the discovery of a S-ERα interaction, imply a role for S as an NRC, and advance knowledge of SARS-CoV-2 biology and coronavirus disease 2019 pathology.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
2021
@article{https://doi.org/10.1111/ejn.15156,
title = {Ventral tegmental area GABA, glutamate, and glutamate-GABA neurons are heterogeneous in their electrophysiological and pharmacological properties},
author = {Jorge Miranda-Barrientos and Ian Chambers and Smriti Mongia and Bing Liu and Hui-Ling Wang and Gabriel E Mateo-Semidey and Elyssa B Margolis and Shiliang Zhang and Marisela Morales},
url = {https://pubmed.ncbi.nlm.nih.gov/33619763/},
doi = {https://doi.org/10.1111/ejn.15156},
year = {2021},
date = {2021-01-01},
urldate = {2021-01-01},
journal = {European Journal of Neuroscience},
volume = {54},
number = {1},
pages = {4061-4084},
abstract = {Abstract The ventral tegmental area (VTA) contains dopamine neurons intermixed with GABA-releasing (expressing vesicular GABA transporter, VGaT), glutamate-releasing (expressing vesicular glutamate transporter 2, VGluT2), and glutamate-GABA co-releasing (co-expressing VGluT2 and VGaT) neurons. By delivering INTRSECT viral vectors into the VTA of double vglut2-Cre/vgat-Flp transgenic mice, we targeted specific VTA cell populations for ex vivo recordings. We found that VGluT2+ VGaT− and VGluT2+ VGaT+ neurons on average had relatively hyperpolarized resting membrane potential, greater rheobase, and lower spontaneous firing frequency compared to VGluT2− VGaT+ neurons, suggesting that VTA glutamate-releasing and glutamate-GABA co-releasing neurons require stronger excitatory drive to fire than GABA-releasing neurons. In addition, we detected expression of Oprm1mRNA (encoding µ opioid receptors, MOR) in VGluT2+ VGaT− and VGluT2− VGaT+ neurons, and that the MOR agonist DAMGO hyperpolarized neurons with these phenotypes. Collectively, we demonstrate the utility of the double transgenic mouse to access VTA glutamate, glutamate-GABA, and GABA neurons to determine their electrophysiological properties. Significant statement Some physiological properties of VTA glutamate-releasing and glutamate-GABA co-releasing neurons are distinct from those of VTA GABA-releasing neurons. µ-opioid receptor activation hyperpolarizes some VTA glutamate-releasing and some GABA-releasing neurons.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{pmid33734555,
title = {NAD^{+} supplementation prevents STING-induced senescence in ataxia telangiectasia by improving mitophagy},
author = {Beimeng Yang and Xiuli Dan and Yujun Hou and Jong-Hyuk Lee and Noah Wechter and Sudarshan Krishnamurthy and Risako Kimura and Mansi Babbar and Tyler Demarest and Ross McDevitt and Shiliang Zhang and Yongqing Zhang and Mark P Mattson and Deborah L Croteau and Vilhelm A Bohr},
url = {https://pubmed.ncbi.nlm.nih.gov/33734555/},
doi = {10.1111/acel.13329},
issn = {1474-9726},
year = {2021},
date = {2021-01-01},
urldate = {2021-01-01},
journal = {Aging Cell},
volume = {20},
number = {4},
pages = {e13329},
abstract = {Senescence phenotypes and mitochondrial dysfunction are implicated in aging and in premature aging diseases, including ataxia telangiectasia (A-T). Loss of mitochondrial function can drive age-related decline in the brain, but little is known about whether improving mitochondrial homeostasis alleviates senescence phenotypes. We demonstrate here that mitochondrial dysfunction and cellular senescence with a senescence-associated secretory phenotype (SASP) occur in A-T patient fibroblasts, and in ATM-deficient cells and mice. Senescence is mediated by stimulator of interferon genes (STING) and involves ectopic cytoplasmic DNA. We further show that boosting intracellular NAD levels with nicotinamide riboside (NR) prevents senescence and SASP by promoting mitophagy in a PINK1-dependent manner. NR treatment also prevents neurodegeneration, suppresses senescence and neuroinflammation, and improves motor function in Atm mice. Our findings suggest a central role for mitochondrial dysfunction-induced senescence in A-T pathogenesis, and that enhancing mitophagy as a potential therapeutic intervention.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
2020
@article{Barbano:2020aa,
title = {VTA Glutamatergic Neurons Mediate Innate Defensive Behaviors},
author = {Flavia M Barbano and Hui-Ling Wang and Shiliang Zhang and Jorge Miranda-Barrientos and David J Estrin and Almaris Figueroa-González and Bing Liu and David J Barker and Marisela Morales},
url = {https://pubmed.ncbi.nlm.nih.gov/32442399/},
doi = {10.1016/j.neuron.2020.04.024},
isbn = {0896-6273},
year = {2020},
date = {2020-07-22},
urldate = {2020-07-22},
booktitle = {Neuron},
journal = {Neuron},
volume = {107},
number = {2},
pages = {368--382.e8},
publisher = {Elsevier},
abstract = {The ventral tegmental area (VTA) has dopamine, GABA, and glutamate neurons, which have been implicated in reward and aversion. Here, we determined whether VTA-glutamate or -GABA neurons play a role in innate defensive behavior. By VTA cell-type-specific genetic ablation, we found that ablation of glutamate, but not GABA, neurons abolishes escape behavior in response to threatening stimuli. We found that escape behavior is also decreased by chemogenetic inhibition of VTA-glutamate neurons and detected increases in activity in VTA-glutamate neurons in response to the threatening stimuli. By ultrastructural and electrophysiological analysis, we established that VTA-glutamate neurons receive a major monosynaptic glutamatergic input from the lateral hypothalamic area (LHA) and found that photoinhibition of this input decreases escape responses to threatening stimuli. These findings indicate that VTA-glutamate neurons are activated by and required for innate defensive responses and that information on threatening stimuli to VTA-glutamate neurons is relayed by LHA-glutamate neurons.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{pmid32877676,
title = {Distinct Signaling by Ventral Tegmental Area Glutamate, GABA, and Combinatorial Glutamate-GABA Neurons in Motivated Behavior},
author = {David H Root and David J Barker and David J Estrin and Jorge A Miranda-Barrientos and Bing Liu and Shiliang Zhang and Hui-Ling Wang and Francois Vautier and Charu Ramakrishnan and Yoon Seok Kim and Lief Fenno and Karl Deisseroth and Marisela Morales},
url = {https://pubmed.ncbi.nlm.nih.gov/32877676/},
doi = {10.1016/j.celrep.2020.108094},
issn = {2211-1247},
year = {2020},
date = {2020-01-01},
urldate = {2020-01-01},
journal = {Cell Rep},
volume = {32},
number = {9},
pages = {108094},
abstract = {Ventral tegmental area (VTA) neurons play roles in reward and aversion. We recently discovered that the VTA has neurons that co-transmit glutamate and GABA (glutamate-GABA co-transmitting neurons), transmit glutamate without GABA (glutamate-transmitting neurons), or transmit GABA without glutamate (GABA-transmitting neurons). However, the functions of these VTA cell types in motivated behavior are unclear. To identify the functions of these VTA cell types, we combine recombinase mouse lines with INTRSECT2.0 vectors to selectively target these neurons. We find that VTA cell types have unique signaling patterns for reward, aversion, and learned cues. Whereas VTA glutamate-transmitting neurons signal cues predicting reward, VTA GABA-transmitting neurons signal cues predicting the absence of reward, and glutamate-GABA co-transmitting neurons signal rewarding and aversive outcomes without signaling learned cues related to those outcomes. Thus, we demonstrate that genetically defined subclasses of VTA glutamate and GABA neurons signal different aspects of motivated behavior.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
2019
@article{Mongia:2019aa,
title = {The Ventral Tegmental Area has calbindin neurons with the capability to co-release glutamate and dopamine into the nucleus accumbens.},
author = {Smriti Mongia and Tsuyoshi Yamaguchi and Bing Liu and Shiliang Zhang and Huiling Wang and Marisela Morales},
url = {https://www.ncbi.nlm.nih.gov/pubmed/31215698},
doi = {10.1111/ejn.14493},
issn = {1460-9568 (Electronic); 0953-816X (Linking)},
year = {2019},
date = {2019-06-19},
urldate = {2019-06-19},
journal = {Eur J Neurosci},
address = {National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA.},
abstract = {The ventral tegmental area (VTA) has three major classes of neurons: dopaminergic (expressing tyrosine hydroxylase; TH), GABAergic (expressing vesicular GABA transporter; VGaT) and glutamatergic (expressing vesicular glutamate transporter 2; VGluT2). While VTA dopaminergic and GABAergic neurons have been further characterized by expression of calcium-binding proteins (calbindin, CB; calretinin, CR or parvalbumin, PV), it is unclear whether these proteins are expressed in rat VTA glutamatergic neurons. Here, by a combination of in situ hybridization (for VGluT2 mRNA detection) and immunohistochemistry (for CB-, CR- or PV-detection), we found that among the total population of VGluT2 neurons, 30% coexpressed CB, 3% coexpressed PV and <1% coexpressed CR. Given that some VGluT2 neurons coexpress TH or VGaT, we examined whether these neurons coexpress CB, and found that about 20% of VGluT2-CB neurons coexpressed TH and about 13% coexpressed VGaT. Because VTA TH-CB neurons are known to target the nucleus accumbens (nAcc), we determined whether VGluT2-CB-TH neurons innervate nAcc, and found that about 80% of VGluT2-CB neurons innervating the nAcc shell coexpressed TH. In summary, (a) CB, PV and CR are detected in subpopulations of VTA-VGluT2 neurons; (b) CB is the main calcium-binding protein present in VTA-VGluT2 neurons; (c) one-third of VTA-VGluT2 neurons coexpress CB; (d) some VTA-VGluT2-CB neurons have the capability to co-release dopamine or GABA, and (e) a subpopulation of VTA glutamatergic-dopaminergic neurons innervates nAcc shell. These findings further provide evidence for molecular diversity among VTA-VGluT2 neurons, neurons that may play a role in specific circuitry and behaviours.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{Zhang2019,
title = {Ultrastructural Detection of Neuronal Markers, Receptors, and Vesicular Transporters},
author = {Shiliang Zhang and Marisela Morales},
doi = {10.1002/cpns.70},
year = {2019},
date = {2019-05-20},
journal = {Current Protocols in Neuroscience},
volume = {88},
number = {1},
abstract = {At the ultrastructural level, axon terminals containing synaptic vesicles are clearly observed. These axon terminals (presynaptic component of a synapse) may be seen establishing contacts (synapses) with cell bodies, axons, or dendrites (postsynaptic component of a synapse). By a combination of ultrastructural analysis and immunodetection of molecules, it is possible to determine the subcellular distribution of specific cellular markers (i.e., enzymes), neurotransmitters (within synaptic vesicles), vesicular transporters (in association with vesicles), and receptors (within the presynaptic or postsynaptic component of a synapse). Here we will provide detailed protocols that facilitate the ultrastructural detection of cellular markers, receptors, and vesicular transporters. These protocols include brain ultrastructural immunodetection of one, two, or three different types of molecules prior to brain tissue processing for ultrastructural analysis (pre‐embedding immunolabeling), brain molecular immunodetection after tissue processing for ultrastructural analysis (post‐embedding immunolabeling), or molecular immunodetection in purified synaptic vesicles. Published 2019. This article is a US Government work and is in the public domain in the USA.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{Zhang:2019aa,
title = {Senolytic therapy alleviates Abeta-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer's disease model.},
author = {Peisu Zhang and Yuki Kishimoto and Ioannis Grammatikakis and Kamalvishnu Gottimukkala and Roy G Cutler and Shiliang Zhang and Kotb Abdelmohsen and Vilhelm A Bohr and Jyoti Misra Sen and Myriam Gorospe and Mark P Mattson},
url = {https://www.ncbi.nlm.nih.gov/pubmed/30936558},
doi = {10.1038/s41593-019-0372-9},
issn = {1546-1726 (Electronic); 1097-6256 (Linking)},
year = {2019},
date = {2019-05-01},
journal = {Nat Neurosci},
volume = {22},
number = {5},
pages = {719--728},
address = {Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD, USA. zhangpei@mail.nih.gov.},
abstract = {Neuritic plaques, a pathological hallmark in Alzheimer's disease (AD) brains, comprise extracellular aggregates of amyloid-beta (Abeta) peptide and degenerating neurites that accumulate autolysosomes. We found that, in the brains of patients with AD and in AD mouse models, Abeta plaque-associated Olig2- and NG2-expressing oligodendrocyte progenitor cells (OPCs), but not astrocytes, microglia, or oligodendrocytes, exhibit a senescence-like phenotype characterized by the upregulation of p21/CDKN1A, p16/INK4/CDKN2A proteins, and senescence-associated beta-galactosidase activity. Molecular interrogation of the Abeta plaque environment revealed elevated levels of transcripts encoding proteins involved in OPC function, replicative senescence, and inflammation. Direct exposure of cultured OPCs to aggregating Abeta triggered cell senescence. Senolytic treatment of AD mice selectively removed senescent cells from the plaque environment, reduced neuroinflammation, lessened Abeta load, and ameliorated cognitive deficits. Our findings suggest a role for Abeta-induced OPC cell senescence in neuroinflammation and cognitive deficits in AD, and a potential therapeutic benefit of senolytic treatments.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{Wang:2019aa,
title = {Dorsal Raphe Dual Serotonin-Glutamate Neurons Drive Reward by Establishing Excitatory Synapses on VTA Mesoaccumbens Dopamine Neurons.},
author = {Hui-Ling Wang and Shiliang Zhang and Jia Qi and Huikun Wang and Roger Cachope and Carlos A Mejias-Aponte and Jorge A Gomez and Gabriel E Mateo-Semidey and Gerard M J Beaudoin and Carlos A Paladini and Joseph F Cheer and Marisela Morales},
url = {https://www.ncbi.nlm.nih.gov/pubmed/30699344},
doi = {10.1016/j.celrep.2019.01.014},
issn = {2211-1247 (Electronic)},
year = {2019},
date = {2019-01-29},
journal = {Cell Rep},
volume = {26},
number = {5},
pages = {1128--1142},
address = {National Institute on Drug Abuse, Neuronal Networks Section, NIH, Baltimore, MD, USA.},
abstract = {Dorsal raphe (DR) serotonin neurons provide a major input to the ventral tegmental area (VTA). Here, we show that DR serotonin transporter (SERT) neurons establish both asymmetric and symmetric synapses on VTA dopamine neurons, but most of these synapses are asymmetric. Moreover, the DR-SERT terminals making asymmetric synapses on VTA dopamine neurons coexpress vesicular glutamate transporter 3 (VGluT3; transporter for accumulation of glutamate for its synaptic release), suggesting the excitatory nature of these synapses. VTA photoactivation of DR-SERT fibers promotes conditioned place preference, elicits excitatory currents on mesoaccumbens dopamine neurons, increases their firing, and evokes dopamine release in nucleus accumbens. These effects are blocked by VTA inactivation of glutamate and serotonin receptors, supporting the idea of glutamate release in VTA from dual DR SERT-VGluT3 inputs. Our findings suggest a path-specific input from DR serotonergic neurons to VTA that promotes reward by the release of glutamate and activation of mesoaccumbens dopamine neurons.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
2018
@article{Root:2018aa,
title = {Selective Brain Distribution and Distinctive Synaptic Architecture of Dual Glutamatergic-GABAergic Neurons.},
author = {David H Root and Shiliang Zhang and David J Barker and Jorge Miranda-Barrientos and Bing Liu and Hui-Ling Wang and Marisela Morales},
url = {https://www.ncbi.nlm.nih.gov/pubmed/29924991},
doi = {10.1016/j.celrep.2018.05.063},
issn = {2211-1247 (Electronic)},
year = {2018},
date = {2018-06-19},
journal = {Cell Rep},
volume = {23},
number = {12},
pages = {3465--3479},
address = {Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Blvd., Suite 200, Baltimore, MD 21224, USA.},
abstract = {For decades, it has been thought that glutamate and GABA are released by distinct neurons. However, some mouse neurons innervating the lateral habenula (LHb) co-release glutamate and GABA. Here, we mapped the distribution of neurons throughout the rat brain that co-express vesicular transporters for the accumulation of glutamate (VGluT2) or GABA (VGaT) and for GABA synthesis (GAD). We found concentrated groups of neurons that co-express VGluT2, VGaT, and GAD mRNAs within subdivisions of the ventral tegmental area (VTA), entopeduncular (EPN), and supramammillary (SUM) nuclei. Single axon terminals established by VTA, EPN, or SUM neurons form a common synaptic architecture involving asymmetric (putative excitatory) and symmetric (putative inhibitory) synapses. Within the LHb, which receives co-transmitted glutamate and GABA from VTA and EPN, VGluT2 and VGaT are distributed on separate synaptic vesicles. We conclude that single axon terminals from VGluT2 and VGaT co-expressing neurons co-transmit glutamate and GABA from distinct synaptic vesicles at independent synapses.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
2017
@article{Mateo:2017aa,
title = {Endocannabinoid Actions on Cortical Terminals Orchestrate Local Modulation of Dopamine Release in the Nucleus Accumbens.},
author = {Yolanda Mateo and Kari A Johnson and Dan P Covey and Brady K Atwood and Hui-Ling Wang and Shiliang Zhang and Iness Gildish and Roger Cachope and Luigi Bellocchio and Manuel Guzman and Marisela Morales and Joseph F Cheer and David M Lovinger},
url = {https://www.ncbi.nlm.nih.gov/pubmed/29216450},
doi = {10.1016/j.neuron.2017.11.012},
issn = {1097-4199 (Electronic); 0896-6273 (Linking)},
year = {2017},
date = {2017-12-06},
journal = {Neuron},
volume = {96},
number = {5},
pages = {1112--1126},
address = {Section on Synaptic Pharmacology, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, Rockville, MD, USA.},
abstract = {Dopamine (DA) transmission mediates numerous aspects of behavior. Although DA release is strongly linked to firing of DA neurons, recent developments indicate the importance of presynaptic modulation at striatal dopaminergic terminals. The endocannabinoid (eCB) system regulates DA release and is a canonical gatekeeper of goal-directed behavior. Here we report that extracellular DA increases induced by selective optogenetic activation of cholinergic neurons in the nucleus accumbens (NAc) are inhibited by CB1 agonists and eCBs. This modulation requires CB1 receptors on cortical glutamatergic afferents. Dopamine increases driven by optogenetic activation of prefrontal cortex (PFC) terminals in the NAc are similarly modulated by activation of these CB1 receptors. We further demonstrate that this same population of CB1 receptors modulates optical self-stimulation sustained by activation of PFC afferents in the NAc. These results establish local eCB actions on PFC terminals within the NAc that inhibit mesolimbic DA release and constrain reward-driven behavior.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{Barker:2017aab,
title = {Lateral Preoptic Control of the Lateral Habenula through Convergent Glutamate and GABA Transmission.},
author = {David J Barker and Jorge Miranda-Barrientos and Shiliang Zhang and David H Root and Hui-Ling Wang and Bing Liu and Erin S Calipari and Marisela Morales},
url = {https://www.ncbi.nlm.nih.gov/pubmed/29141211},
doi = {10.1016/j.celrep.2017.10.066},
issn = {2211-1247 (Electronic)},
year = {2017},
date = {2017-11-14},
journal = {Cell Rep},
volume = {21},
number = {7},
pages = {1757--1769},
address = {Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Blvd., Suite 200, Baltimore, MD 21224, USA.},
abstract = {The lateral habenula (LHb) is a brain structure that participates in cognitive and emotional processing and has been implicated in several mental disorders. Although one of the largest inputs to the LHb originates in the lateral preoptic area (LPO), little is known about how the LPO participates in the regulation of LHb function. Here, we provide evidence that the LPO exerts bivalent control over the LHb through the convergent transmission of LPO glutamate and gamma-aminobutyric acid (GABA) onto single LHb neurons. In vivo, both LPO-glutamatergic and LPO-GABAergic inputs to the LHb are activated by aversive stimuli, and their predictive cues yet produce opposing behaviors when stimulated independently. These results support a model wherein the balanced response of converging LPO-glutamate and LPO-GABA are necessary for a normal response to noxious stimuli, and an imbalance in LPO-->LHb glutamate or GABA results in the type of aberrant processing that may underlie mental disorders.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{Venniro2017,
title = {The Anterior Insular Cortex-->Central Amygdala Glutamatergic Pathway Is Critical to Relapse after Contingency Management.},
author = {Marco Venniro and Daniele Caprioli and Michelle Zhang and Leslie R Whitaker and Shiliang Zhang and Brandon L Warren and Carlo Cifani and Nathan J Marchant and Ofer Yizhar and Jennifer M Bossert and Cristiano Chiamulera and Marisela Morales and Yavin Shaham},
url = {https://www.ncbi.nlm.nih.gov/pubmed/29024664},
doi = {10.1016/j.neuron.2017.09.024},
issn = {1097-4199 (Electronic); 0896-6273 (Linking)},
year = {2017},
date = {2017-10-11},
urldate = {2017-10-11},
journal = {Neuron},
volume = {96},
number = {2},
pages = {414--427},
address = {Behavioral Neuroscience Research Branch, Intramural Research Program, NIDA, NIH, Baltimore, MD, USA. Electronic address: venniro.marco@nih.gov.},
abstract = {Despite decades of research on neurobiological mechanisms of psychostimulant addiction, the only effective treatment for many addicts is contingency management, a behavioral treatment that uses alternative non-drug reward to maintain abstinence. However, when contingency management is discontinued, most addicts relapse to drug use. The brain mechanisms underlying relapse after cessation of contingency management are largely unknown, and, until recently, an animal model of this human condition did not exist. Here we used a novel rat model, in which the availability of a mutually exclusive palatable food maintains prolonged voluntary abstinence from intravenous methamphetamine self-administration, to demonstrate that the activation of monosynaptic glutamatergic projections from anterior insular cortex to central amygdala is critical to relapse after the cessation of contingency management. We identified the anterior insular cortex-to-central amygdala projection as a new addiction- and motivation-related projection and a potential target for relapse prevention.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{Edwards:2017aa,
title = {Circuit specificity in the inhibitory architecture of the VTA regulates cocaine-induced behavior.},
author = {Nicholas J Edwards and Hugo A Tejeda and Marco Pignatelli and Shiliang Zhang and Ross A McDevitt and Jocelyn Wu and Caroline E Bass and Bernhard Bettler and Marisela Morales and Antonello Bonci},
url = {https://www.ncbi.nlm.nih.gov/pubmed/28114294},
doi = {10.1038/nn.4482},
issn = {1546-1726 (Electronic); 1097-6256 (Linking)},
year = {2017},
date = {2017-03-01},
journal = {Nat Neurosci},
volume = {20},
number = {3},
pages = {438--448},
address = {Intramural Research Program, National Institute on Drug Abuse, US National Institutes of Health, Baltimore, Maryland, USA.},
abstract = {Afferent inputs to the ventral tegmental area (VTA) control reward-related behaviors through regulation of dopamine neuron activity. The nucleus accumbens (NAc) provides one of the most prominent projections to the VTA; however, recent studies have provided conflicting evidence regarding the function of these inhibitory inputs. Using optogenetics, cell-specific ablation, whole cell patch-clamp and immuno-electron microscopy, we found that NAc inputs synapsed directly onto dopamine neurons, preferentially activating GABAB receptors. GABAergic inputs from the NAc and local VTA GABA neurons were differentially modulated and activated separate receptor populations in dopamine neurons. Genetic deletion of GABAB receptors from dopamine neurons in adult mice did not affect general or morphine-induced locomotor activity, but markedly increased cocaine-induced locomotion. Collectively, our findings demonstrate notable selectivity in the inhibitory architecture of the VTA and suggest that long-range GABAergic inputs to dopamine neurons fundamentally regulate behavioral responses to cocaine.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{Steidl:2017aa,
title = {Optogenetic excitation in the ventral tegmental area of glutamatergic or cholinergic inputs from the laterodorsal tegmental area drives reward.},
author = {Stephan Steidl and Huiling Wang and Marco Ordonez and Shiliang Zhang and Marisela Morales},
url = {https://www.ncbi.nlm.nih.gov/pubmed/27740714},
doi = {10.1111/ejn.13436},
issn = {1460-9568 (Electronic); 0953-816X (Linking)},
year = {2017},
date = {2017-02-01},
journal = {Eur J Neurosci},
volume = {45},
number = {4},
pages = {559--571},
address = {Department of Psychology, Loyola University Chicago, 1032 West Sheridan Road, Chicago, IL, 60626, USA.},
abstract = {Converging evidence shows that ventral tegmental area (VTA) dopamine neurons receive laterodorsal tegmental nucleus (LDTg) cholinergic and glutamatergic inputs. To test the behavioral consequences of selectively driving the two sources of excitatory LDTg input to the VTA, channelrhodopsin-2 (ChR2) was expressed in LDTg cholinergic neurons of ChAT::Cre mice (ChAT-ChR2 mice) or in LDTg glutamatergic neurons of VGluT2::Cre mice (VGluT2-ChR2 mice). Mice were tested in a 3-chamber place preference apparatus where entry into a light-paired chamber resulted in VTA light stimulation of LDTg-cholinergic or LDTg-glutamatergic axons for the duration of a chamber stay. ChAT-ChR2 mice spent more time in the light-paired chamber and subsequently showed conditioned place preference for the light-paired chamber in the absence of light. VGluT2-ChR2 mice, entered the light-paired chamber significantly more times than a light-unpaired chamber, but remained in the light-paired chamber for short time periods and did not show a conditioned place preference. When each entry into the light-paired chamber resulted in a single train of VTA light stimulation, VGluT2-ChR2 mice entered the light-paired chamber significantly more times than the light-unpaired chamber, but spent approximately equal amounts of time in the two chambers. VTA excitation of LDTg-glutamatergic inputs may be more important for reinforcement of initial chamber entry while VTA excitation of LDTg-cholinergic inputs may be more important for the rewarding effects of chamber stays. We suggest that LDTg-cholinergic and LDTg-glutamatergic inputs to the VTA each contribute to the net rewarding effects of exciting LDTg axons in the VTA.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Confocal and Electron Microscopy Core – Publications
Publications from the Electron Microscopy Core. Tdrd3-null mice show post-transcriptional and behavioral impairments associated with neurogenesis and synaptic plasticity Journal Article In: Prog Neurobiol, vol. 233, pp. 102568, 2024, ISSN: 1873-5118. Lateral hypothalamic glutamatergic inputs to VTA glutamatergic neurons mediate prioritization of innate defensive behavior over feeding Journal Article In: Nat Commun, vol. 15, no. 1, pp. 403, 2024, ISSN: 2041-1723. Lateral preoptic area glutamate neurons relay nociceptive information to the ventral tegmental area Journal Article In: Cell Rep, vol. 42, no. 9, pp. 113029, 2023, ISSN: 2211-1247. Distinct sub-second dopamine signaling in dorsolateral striatum measured by a genetically-encoded fluorescent sensor Journal Article In: Nat Commun, vol. 14, no. 1, pp. 5915, 2023, ISSN: 2041-1723. Expression of the excitatory opsin ChRERα can be traced longitudinally in rat and nonhuman primate brains with PET imaging Journal Article In: Sci Transl Med, vol. 15, no. 706, pp. eadd1014, 2023, ISSN: 1946-6242. In: Microsc Microanal, vol. 29, no. 29 Suppl 1, pp. 1192–1194, 2023, ISSN: 1435-8115. In: J Neurosci, vol. 43, no. 23, pp. 4217–4233, 2023, ISSN: 1529-2401. The SARS-CoV-2 spike protein binds and modulates estrogen receptors Journal Article In: Sci Adv, vol. 8, no. 48, pp. eadd4150, 2022, ISSN: 2375-2548. In: European Journal of Neuroscience, vol. 54, no. 1, pp. 4061-4084, 2021. NAD+ supplementation prevents STING-induced senescence in ataxia telangiectasia by improving mitophagy Journal Article In: Aging Cell, vol. 20, no. 4, pp. e13329, 2021, ISSN: 1474-9726. VTA Glutamatergic Neurons Mediate Innate Defensive Behaviors Journal Article In: Neuron, vol. 107, no. 2, pp. 368–382.e8, 2020, ISBN: 0896-6273. Distinct Signaling by Ventral Tegmental Area Glutamate, GABA, and Combinatorial Glutamate-GABA Neurons in Motivated Behavior Journal Article In: Cell Rep, vol. 32, no. 9, pp. 108094, 2020, ISSN: 2211-1247. The Ventral Tegmental Area has calbindin neurons with the capability to co-release glutamate and dopamine into the nucleus accumbens. Journal Article In: Eur J Neurosci, 2019, ISSN: 1460-9568 (Electronic); 0953-816X (Linking). Ultrastructural Detection of Neuronal Markers, Receptors, and Vesicular Transporters Journal Article In: Current Protocols in Neuroscience, vol. 88, no. 1, 2019. Senolytic therapy alleviates Abeta-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer's disease model. Journal Article In: Nat Neurosci, vol. 22, no. 5, pp. 719–728, 2019, ISSN: 1546-1726 (Electronic); 1097-6256 (Linking). Dorsal Raphe Dual Serotonin-Glutamate Neurons Drive Reward by Establishing Excitatory Synapses on VTA Mesoaccumbens Dopamine Neurons. Journal Article In: Cell Rep, vol. 26, no. 5, pp. 1128–1142, 2019, ISSN: 2211-1247 (Electronic). Selective Brain Distribution and Distinctive Synaptic Architecture of Dual Glutamatergic-GABAergic Neurons. Journal Article In: Cell Rep, vol. 23, no. 12, pp. 3465–3479, 2018, ISSN: 2211-1247 (Electronic). Endocannabinoid Actions on Cortical Terminals Orchestrate Local Modulation of Dopamine Release in the Nucleus Accumbens. Journal Article In: Neuron, vol. 96, no. 5, pp. 1112–1126, 2017, ISSN: 1097-4199 (Electronic); 0896-6273 (Linking). Lateral Preoptic Control of the Lateral Habenula through Convergent Glutamate and GABA Transmission. Journal Article In: Cell Rep, vol. 21, no. 7, pp. 1757–1769, 2017, ISSN: 2211-1247 (Electronic). The Anterior Insular Cortex-->Central Amygdala Glutamatergic Pathway Is Critical to Relapse after Contingency Management. Journal Article In: Neuron, vol. 96, no. 2, pp. 414–427, 2017, ISSN: 1097-4199 (Electronic); 0896-6273 (Linking). Circuit specificity in the inhibitory architecture of the VTA regulates cocaine-induced behavior. Journal Article In: Nat Neurosci, vol. 20, no. 3, pp. 438–448, 2017, ISSN: 1546-1726 (Electronic); 1097-6256 (Linking). Optogenetic excitation in the ventral tegmental area of glutamatergic or cholinergic inputs from the laterodorsal tegmental area drives reward. Journal Article In: Eur J Neurosci, vol. 45, no. 4, pp. 559–571, 2017, ISSN: 1460-9568 (Electronic); 0953-816X (Linking).
2024
2023
2022
2021
2020
2019
2018
2017