161 results on '"Francis S, Lee"'
Search Results
2. Regulation of social interaction in mice by a frontostriatal circuit modulated by established hierarchical relationships
- Author
-
Robert N. Fetcho, Baila S. Hall, David J. Estrin, Alexander P. Walsh, Peter J. Schuette, Jesse Kaminsky, Ashna Singh, Jacob Roshgodal, Charlotte C. Bavley, Viraj Nadkarni, Susan Antigua, Thu N. Huynh, Logan Grosenick, Camille Carthy, Lauren Komer, Avishek Adhikari, Francis S. Lee, Anjali M. Rajadhyaksha, and Conor Liston
- Subjects
Multidisciplinary ,General Physics and Astronomy ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology - Abstract
Social hierarchies exert a powerful influence on behavior, but the neurobiological mechanisms that detect and regulate hierarchical interactions are not well understood, especially at the level of neural circuits. Here, we use fiber photometry and chemogenetic tools to record and manipulate the activity of nucleus accumbens-projecting cells in the ventromedial prefrontal cortex (vmPFC-NAcSh) during tube test social competitions. We show that vmPFC-NAcSh projections signal learned hierarchical relationships, and are selectively recruited by subordinate mice when they initiate effortful social dominance behavior during encounters with a dominant competitor from an established hierarchy. After repeated bouts of social defeat stress, this circuit is preferentially activated during social interactions initiated by stress resilient individuals, and plays a necessary role in supporting social approach behavior in subordinated mice. These results define a necessary role for vmPFC-NAcSh cells in the adaptive regulation of social interaction behavior based on prior hierarchical interactions.
- Published
- 2023
- Full Text
- View/download PDF
3. Neurotransmission-related gene expression in the frontal pole is altered in subjects with bipolar disorder and schizophrenia
- Author
-
Adriana M. Medina, Megan Hastings Hagenauer, David M. Krolewski, Evan Hughes, Liam Cannon Thew Forrester, David M. Walsh, Maria Waselus, Evelyn Richardson, Cortney A. Turner, P. Adolfo Sequeira, Preston M. Cartagena, Robert C. Thompson, Marquis P. Vawter, Blynn G. Bunney, Richard M. Myers, Jack D. Barchas, Francis S. Lee, Alan F. Schatzberg, William E. Bunney, Huda Akil, and Stanley J. Watson
- Subjects
Bipolar Disorder ,Adolescent ,Clinical Sciences ,Neurosciences ,Gene Expression ,Serious Mental Illness ,Synaptic Transmission ,Frontal Lobe ,Brain Disorders ,Cellular and Molecular Neuroscience ,Psychiatry and Mental health ,Mental Health ,Good Health and Well Being ,Schizophrenia ,Genetics ,Public Health and Health Services ,Humans ,2.1 Biological and endogenous factors ,Psychology ,Aetiology ,Biological Psychiatry - Abstract
The frontal pole (Brodmann area 10, BA10) is the largest cytoarchitectonic region of the human cortex, performing complex integrative functions. BA10 undergoes intensive adolescent grey matter pruning prior to the age of onset for bipolar disorder (BP) and schizophrenia (SCHIZ), and its dysfunction is likely to underly aspects of their shared symptomology. In this study, we investigated the role of BA10 neurotransmission-related gene expression in BP and SCHIZ. We performed qPCR to measure the expression of 115 neurotransmission-related targets in control, BP, and SCHIZ postmortem samples (n = 72). We chose this method for its high sensitivity to detect low-level expression. We then strengthened our findings by performing a meta-analysis of publicly released BA10 microarray data (n = 101) and identified sources of convergence with our qPCR results. To improve interpretation, we leveraged the unusually large database of clinical metadata accompanying our samples to explore the relationship between BA10 gene expression, therapeutics, substances of abuse, and symptom profiles, and validated these findings with publicly available datasets. Using these convergent sources of evidence, we identified 20 neurotransmission-related genes that were differentially expressed in BP and SCHIZ in BA10. These results included a large diagnosis-related decrease in two important therapeutic targets with low levels of expression, HTR2B and DRD4, as well as other findings related to dopaminergic, GABAergic and astrocytic function. We also observed that therapeutics may produce a differential expression that opposes diagnosis effects. In contrast, substances of abuse showed similar effects on BA10 gene expression as BP and SCHIZ, potentially amplifying diagnosis-related dysregulation.
- Published
- 2023
4. Large Common Mitochondrial DNA Deletions Are Associated with a Mitochondrial SNP T14798C Near the 3′ Breakpoints
- Author
-
Brooke E. Hjelm, Christian Ramiro, Brandi L. Rollins, Audrey A. Omidsalar, Daniel S. Gerke, Sujan C. Das, Adolfo Sequeira, Ling Morgan, Alan F. Schatzberg, Jack D. Barchas, Francis S. Lee, Richard M. Myers, Stanley J. Watson, Huda Akil, William E. Bunney, and Marquis P. Vawter
- Subjects
General Medicine - Abstract
Introduction: Large somatic deletions of mitochondrial DNA (mtDNA) accumulate with aging in metabolically active tissues such as the brain. We have cataloged the breakpoints and frequencies of large mtDNA deletions in the human brain. Methods: We quantified 112 high-frequency mtDNA somatic deletions across four human brain regions with the Splice-Break2 pipeline. In addition, we utilized PLINK/Seq to test the association of mitochondrial genotypes with the abundance of these high-frequency mtDNA deletions. A conservative p value threshold of 5E−08 was used to find the significant loci. Results: One mtDNA SNP (T14798C) was significantly associated with mtDNA deletions in two brain regions, the dorsolateral prefrontal cortex (DLPFC) and the superior temporal gyrus. Since the DLPFC showed the most robust association between T14798C and two deletion breakpoints (7816–14807 and 5462–14807), this association was tested in the DLPFC of a replication sample and validated the first results. Incorporating the C allele at 14,798 bp increased the perfect/imperfect length of the repeat at the 3′ breakpoint of the two associated deletions. Conclusion: This is the first study to identify the association of mtDNA SNP with large mtDNA deletions in the human brain. The T14798C allele located in the MT-CYB gene is a common polymorphism that occurs in several mitochondrial haplogroups. We hypothesize that the T14798C association with two deletions occurs by extending the repeat length around the 3′ deletion breakpoints. This simple mechanism suggests that mtDNA SNPs can affect the mitochondrial genome structure, especially in brain where high levels of reactive oxygen species lead to deletion accumulation with aging.
- Published
- 2022
- Full Text
- View/download PDF
5. Genomic modules and intramodular network concordance in susceptible and resilient male mice across models of stress
- Author
-
Nathan R. Einhorn, Xianglan Wen, Huda Akil, Bruce S. McEwen, Francis S. Lee, Nicholas O’Toole, Michael J. Meaney, Jordan Marrocco, Mojun Shen, Carine Parent, Tie-Yuan Zhang, Salvatore G. Caradonna, and Huzefa Khalil
- Subjects
Pharmacology ,Genetics ,Environmental enrichment ,education.field_of_study ,Concordance ,Population ,Gene regulatory network ,Biology ,Social defeat ,Psychiatry and Mental health ,Neurotrophic factors ,Genetic predisposition ,education ,Gene - Abstract
The multifactorial etiology of stress-related disorders necessitates a constant interrogation of the molecular convergences in preclinical models of stress that use disparate paradigms as stressors spanning from environmental challenges to genetic predisposition to hormonal signaling. Using RNA-sequencing, we investigated the genomic signatures in the ventral hippocampus common to mouse models of stress. Chronic oral corticosterone (CORT) induced increased anxiety- and depression-like behavior in wild-type male mice and male mice heterozygous for the gene coding for brain-derived neurotrophic factor Val66Met, a variant associated with genetic susceptibility to stress. In a separate set of male mice, chronic social defeat stress (CSDS) led to a susceptible or a resilient population, whose proportion was dependent on housing conditions, namely standard housing or enriched environment. Rank-rank-hypergeometric overlap (RRHO), a threshold-free approach that ranks genes by their p value and effect size direction, was used to identify genes from a continuous gradient of significancy that were concordant across groups. In mice treated with CORT and in standard-housed susceptible mice, differentially expressed genes (DEGs) were concordant for gene networks involved in neurotransmission, cytoskeleton function, and vascularization. Weighted gene co-expression analysis generated 54 gene hub modules and revealed two modules in which both CORT and CSDS-induced enrichment in DEGs, whose function was concordant with the RRHO predictions, and correlated with behavioral resilience or susceptibility. These data showed transcriptional concordance across models in which the stress coping depends upon hormonal, environmental, or genetic factors revealing common genomic drivers that embody the multifaceted nature of stress-related disorders.
- Published
- 2021
- Full Text
- View/download PDF
6. Author response: A genetic variant of fatty acid amide hydrolase (FAAH) exacerbates hormone-mediated orexigenic feeding in mice
- Author
-
Georgia Balsevich, Gavin N Petrie, Daniel E Heinz, Arashdeep Singh, Robert J Aukema, Avery C Hunker, Haley A Vecchiarelli, Hiulan Yau, Martin Sticht, Roger J Thompson, Francis S Lee, Larry S Zweifel, Prasanth K Chelikani, Nils C Gassen, and Matthew N Hill
- Published
- 2022
- Full Text
- View/download PDF
7. Control of Gα
- Author
-
Guoqing, Xiang, Amanda, Acosta-Ruiz, Arthur, Radoux-Mergault, Melanie, Kristt, Jihye, Kim, Jared D, Moon, Johannes, Broichhagen, Asuka, Inoue, Francis S, Lee, Miriam, Stoeber, Jeremy S, Dittman, and Joshua, Levitz
- Abstract
Numerous processes contribute to the regulation of G protein-coupled receptors (GPCRs), but relatively little is known about rapid mechanisms that control signaling on the seconds time scale or regulate cross-talk between receptors. Here, we reveal that the ability of some GPCR kinases (GRKs) to bind Gα
- Published
- 2022
8. Control of Gα q signaling dynamics and GPCR cross-talk by GRKs
- Author
-
Guoqing Xiang, Amanda Acosta-Ruiz, Arthur Radoux-Mergault, Melanie Kristt, Jihye Kim, Jared D. Moon, Johannes Broichhagen, Asuka Inoue, Francis S. Lee, Miriam Stoeber, Jeremy S. Dittman, and Joshua Levitz
- Subjects
Multidisciplinary - Abstract
Numerous processes contribute to the regulation of G protein–coupled receptors (GPCRs), but relatively little is known about rapid mechanisms that control signaling on the seconds time scale or regulate cross-talk between receptors. Here, we reveal that the ability of some GPCR kinases (GRKs) to bind Gα q both drives acute signaling desensitization and regulates functional interactions between GPCRs. GRK2/3-mediated acute desensitization occurs within seconds, is rapidly reversible, and can occur upon local, subcellular activation. This rapid desensitization is kinase independent, insensitive to pharmacological inhibition, and generalizable across receptor families and effectors. We also find that the ability of GRK2 to bind G proteins also enables it to regulate the extent and timing of Gα q -dependent signaling cross-talk between GPCRs. Last, we find that G protein/GRK2 interactions enable a novel form of GPCR trafficking cross-talk. Together, this work reveals potent forms of Gα q -dependent GPCR regulation with wide-ranging pharmacological and physiological implications.
- Published
- 2022
- Full Text
- View/download PDF
9. Overexpression of Cancer- and Neurotransmitter-Associated Genes in the Nucleus Accumbens of Smokers
- Author
-
Richard Stein, Firoza Mamdani, Blynn Bunney, Preston Cartagena, Marquis P. Vawter, Alan F. Schatzberg, Jack Barchas, Francis S. Lee, Richard Myers, Stanley J. Watson, Huda Akil, William E. Bunney, and Adolfo Sequeira
- Abstract
The effects of smoking in the human brain were explored at the molecular level in the nucleus accumbens(NAcc), a key brain region involved in tobacco addiction. Gene expression data from post-mortem NAcc were analyzed according to smoking habits: Never smokers, Former smokers and Current smokers at the time of death. The effect of smoking was determined using an ANCOVA model, controlling for potential confounders (psychiatric diagnosis, gender, age, post-mortem interval, and brain pH) followed by pair-wise post-hoc comparisons. Q-values (false discovery rate adjusted p-values) < 0.05 were used in combination with a fold change of > ±1.3 to identify the most relevant genes. The greatest number of differentially expressed genes (DEGs) were found in subjects with a recent history of smoking (Current smokers) compared to either Former or Never smokers. Only two genes were differentially expressed between Former and Never smokers, suggesting that the effects of smoking on gene expression in the brain may be transient. Ingenuity Pathway Analysis (IPA) of DEGs identified a significant over-representation of neurotransmitter system genes (glutamate, GABA) in Current smokers. IPA also revealed many genes associated with cancer in Current smokers compared to Former and Never smokers despite no known cancer in any subjects. Genes associated with neoplasms, glioblastoma, gliomas and tumor regulations are among the top 10 transcripts.Our findings show that active smokers have a significant increase in cancer-related genes and alterations in glutamate and GABA neurotransmitter systems in the NAcc. To our knowledge this is the first study to identify cancer-related genes in the NAcc in Current smokers who have no evidence of cancer.
- Published
- 2022
- Full Text
- View/download PDF
10. Enhancing exposure therapy for posttraumatic stress disorder (PTSD): a randomized clinical trial of virtual reality and imaginal exposure with a cognitive enhancer
- Author
-
JoAnn Difede, Barbara O. Rothbaum, Albert A. Rizzo, Katarzyna Wyka, Lisa Spielman, Christopher Reist, Michael J. Roy, Tanja Jovanovic, Seth D. Norrholm, Judith Cukor, Megan Olden, Charles E. Glatt, and Francis S. Lee
- Subjects
Stress Disorders, Post-Traumatic ,Cellular and Molecular Neuroscience ,Psychiatry and Mental health ,Treatment Outcome ,Cycloserine ,Brain-Derived Neurotrophic Factor ,Virtual Reality ,Humans ,Implosive Therapy ,Nootropic Agents ,Biological Psychiatry - Abstract
Posttraumatic stress disorder (PTSD) is a significant public health issue. Yet, there are limited treatment options and no data to suggest which treatment will work for whom. We tested the efficacy of virtual reality exposure (VRE) or prolonged imaginal exposure (PE), augmented with D-cycloserine (DCS) for combat-related PTSD. As an exploratory aim, we examined whether brain-derived neurotrophic factor (BDNF) and fatty acid amide hydrolase (FAAH) moderated treatment response. Military personnel with PTSD (n = 192) were recruited into a multisite double-blind randomized controlled trial to receive nine weeks of VRE or PE, with DCS or placebo. Primary outcome was the improvement in symptom severity. Randomization was stratified by comorbid depression (MDD) and site. Participants in both VRE and PE showed similar meaningful clinical improvement with no difference between the treatment groups. A significant interaction (p = 0.45) suggested VRE was more effective for depressed participants (CAPS difference M = 3.51 [95% CI 1.17–5.86], p = 0.004, ES = 0.14) while PE was more effective for nondepressed participants (M = −8.87 [95% CI −11.33 to −6.40], p p = 0.073) suggested that depressed participants improved more on placebo (M = −8.43 [95% CI −10.98 to −5.88], p ClinicalTrials.gov Identifier: NCT01352637.
- Published
- 2022
- Full Text
- View/download PDF
11. An Adolescent Sensitive Period for Threat Responding: Impacts of Stress and Sex
- Author
-
Danielle M. Gerhard, Francis S. Lee, and Heidi C. Meyer
- Subjects
0301 basic medicine ,Adolescent ,Period (gene) ,Affect (psychology) ,Article ,Extinction, Psychological ,Developmental psychology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Pediatric anxiety ,Memory ,Sensitive periods ,Stress (linguistics) ,medicine ,Animals ,Humans ,Learning ,Child ,Biological Psychiatry ,Fear ,Extinction (psychology) ,Biological sex ,Anxiety Disorders ,Rats ,030104 developmental biology ,Anxiety ,medicine.symptom ,Psychology ,030217 neurology & neurosurgery - Abstract
Anxiety and fear-related disorders peak in prevalence during adolescence, a window of rapid behavioral development and neural remodeling. However, understanding of the development of threat responding and the underlying neural circuits remains limited. Preclinical models of threat conditioning and extinction have provided an unparalleled glimpse into the developing brain. In this review we discuss mouse and rat studies on the development of threat response regulation with a focus on the adolescent period. Evidence of non-linear patterns of threat responding during adolescence and the continued development of the underlying circuitry is highly indicative of an adolescent sensitive period for threat response regulation. While we highlight literature in support of this unique developmental window, we also emphasize the need for causal studies to clarify the parameters defining such a sensitive period. In doing so, we explore how stress and biological sex impact the development and expression of threat response regulation during adolescence and beyond. Ultimately, a deeper understanding of how these factors interact with and impact developmental trajectories of learning and memory will inform treatment and prevention strategies for pediatric anxiety disorders.
- Published
- 2021
- Full Text
- View/download PDF
12. A Common Human Brain-Derived Neurotrophic Factor Polymorphism Leads to Prolonged Depression of Excitatory Synaptic Transmission by Isoflurane in Hippocampal Cultures
- Author
-
Riley A, Williams, Kenneth W, Johnson, Francis S, Lee, Hugh C, Hemmings, and Jimcy, Platholi
- Subjects
Cellular and Molecular Neuroscience ,Molecular Biology - Abstract
Multiple presynaptic and postsynaptic targets have been identified for the reversible neurophysiological effects of general anesthetics on synaptic transmission and neuronal excitability. However, the synaptic mechanisms involved in persistent depression of synaptic transmission resulting in more prolonged neurological dysfunction following anesthesia are less clear. Here, we show that brain-derived neurotrophic factor (BDNF), a growth factor implicated in synaptic plasticity and dysfunction, enhances glutamate synaptic vesicle exocytosis, and that attenuation of vesicular BDNF release by isoflurane contributes to transient depression of excitatory synaptic transmission in mice. This reduction in synaptic vesicle exocytosis by isoflurane was acutely irreversible in neurons that release less endogenous BDNF due to a polymorphism (BDNF Val66Met; rs6265) compared to neurons from wild-type mice. These effects were prevented by exogenous application of BDNF. Our findings identify a role for a common human BDNF single nucleotide polymorphism in persistent changes of synaptic function following isoflurane exposure. These short-term persistent alterations in excitatory synaptic transmission indicate a role for human genetic variation in anesthetic effects on synaptic plasticity and neurocognitive function.
- Published
- 2022
- Full Text
- View/download PDF
13. Identification of potential blood biomarkers associated with suicide in major depressive disorder
- Author
-
Firoza Mamdani, Matthieu D. Weber, Blynn Bunney, Kathleen Burke, Preston Cartagena, David Walsh, Francis S. Lee, Jack Barchas, Alan F. Schatzberg, Richard M. Myers, Stanley J. Watson, Huda Akil, Marquis P. Vawter, William E. Bunney, and Adolfo Sequeira
- Subjects
Depressive Disorder, Major ,Suicide ,Cellular and Molecular Neuroscience ,Psychiatry and Mental health ,Amino Acid Transport Systems ,Calcium-Binding Proteins ,mental disorders ,Brain ,Humans ,Prefrontal Cortex ,behavioral disciplines and activities ,Biomarkers ,Biological Psychiatry - Abstract
Suicides have increased to over 48,000 deaths yearly in the United States. Major depressive disorder (MDD) is the most common diagnosis among suicides, and identifying those at the highest risk for suicide is a pressing challenge. The objective of this study is to identify changes in gene expression associated with suicide in brain and blood for the development of biomarkers for suicide. Blood and brain were available for 45 subjects (53 blood samples and 69 dorsolateral prefrontal cortex (DLPFC) samples in total). Samples were collected from MDD patients who died by suicide (MDD-S), MDDs who died by other means (MDD-NS) and non-psychiatric controls. We analyzed gene expression using RNA and the NanoString platform. In blood, we identified 14 genes which significantly differentiated MDD-S versus MDD-NS. The top six genes differentially expressed in blood were: PER3, MTPAP, SLC25A26, CD19, SOX9, and GAR1. Additionally, four genes showed significant changes in brain and blood between MDD-S and MDD-NS; SOX9 was decreased and PER3 was increased in MDD-S in both tissues, while CD19 and TERF1 were increased in blood but decreased in DLPFC. To our knowledge, this is the first study to analyze matched blood and brain samples in a well-defined population of MDDs demonstrating significant differences in gene expression associated with completed suicide. Our results strongly suggest that blood gene expression is highly informative to understand molecular changes in suicide. Developing a suicide biomarker signature in blood could help health care professionals to identify subjects at high risk for suicide.
- Published
- 2022
- Full Text
- View/download PDF
14. mGreenLantern: a bright monomeric fluorescent protein with rapid expression and cell filling properties for neuronal imaging
- Author
-
Hirofumi Morishita, Conor Liston, Mitchell H. Murdock, Cristina Lao-Peregrin, Gregory A. Petsko, Francis S. Lee, Elisa M. Nabel, Benjamin C. Campbell, Pantelis Tsoulfas, and Murray Blackmore
- Subjects
Dendritic spine ,Confocal ,Green Fluorescent Proteins ,Cell ,Fluorescent Antibody Technique ,Gene Expression ,GFP ,Green fluorescent protein ,Mice ,Genes, Reporter ,In vivo ,Microscopy ,medicine ,fluorescent protein ,Animals ,Neurons ,Multidisciplinary ,Protein Stability ,Chemistry ,Spectrum Analysis ,neurobiology ,imaging ,Brain ,protein engineering ,Protein engineering ,Biological Sciences ,Fluorescence ,Molecular Imaging ,Biophysics and Computational Biology ,medicine.anatomical_structure ,Microscopy, Fluorescence ,Solubility ,Physical Sciences ,Mutation ,Proteolysis ,Biophysics ,Neuroscience - Abstract
Significance We have developed a fluorescent protein, mGreenLantern, that features exceptionally high brightness in mouse, bacterial, and human cells (up to sixfold brighter than EGFP) and have demonstrated its superior ability to highlight neuronal morphology compared to EGFP and EYFP. Screening fluorescent protein mutants based on whole-cell brightness while evaluating expression kinetics in lysate enabled us to identify variants exhibiting striking divergences between their computed spectroscopic brightness and actual performance in cells. mGreenLantern additionally features unusually high chemical and thermodynamic stability and is compatible with existing GFP filter sets, excitation sources, commercial EGFP antibodies, expansion microscopy, and whole-brain tissue clearing. Our hypothesis-driven engineering strategy represents a generalizable method with great potential to enhance the performance of constitutive reporters and GFP-based biosensors., Although ubiquitous in biological studies, the enhanced green and yellow fluorescent proteins (EGFP and EYFP) were not specifically optimized for neuroscience, and their underwhelming brightness and slow expression in brain tissue limits the fidelity of dendritic spine analysis and other indispensable techniques for studying neurodevelopment and plasticity. We hypothesized that EGFP’s low solubility in mammalian systems must limit the total fluorescence output of whole cells, and that improving folding efficiency could therefore translate into greater brightness of expressing neurons. By introducing rationally selected combinations of folding-enhancing mutations into GFP templates and screening for brightness and expression rate in human cells, we developed mGreenLantern, a fluorescent protein having up to sixfold greater brightness in cells than EGFP. mGreenLantern illuminates neurons in the mouse brain within 72 h, dramatically reducing lag time between viral transduction and imaging, while its high brightness improves detection of neuronal morphology using widefield, confocal, and two-photon microscopy. When virally expressed to projection neurons in vivo, mGreenLantern fluorescence developed four times faster than EYFP and highlighted long-range processes that were poorly detectable in EYFP-labeled cells. Additionally, mGreenLantern retains strong fluorescence after tissue clearing and expansion microscopy, thereby facilitating superresolution and whole-brain imaging without immunohistochemistry. mGreenLantern can directly replace EGFP/EYFP in diverse systems due to its compatibility with GFP filter sets, recognition by EGFP antibodies, and excellent performance in mouse, human, and bacterial cells. Our screening and rational engineering approach is broadly applicable and suggests that greater potential of fluorescent proteins, including biosensors, could be unlocked using a similar strategy.
- Published
- 2020
- Full Text
- View/download PDF
15. Zinc induced structural changes in the intrinsically disordered BDNF Met prodomain confer synaptic elimination
- Author
-
Thomas A. Neubert, William J. Rice, Barbara L. Hempstead, Jing Wang, David G Clossey, Henrietta Bains, Clay Bracken, Jingjing Deng, Agustin Anastasia, Francis S. Lee, and Joanna Giza
- Subjects
0301 basic medicine ,Conformational change ,Magnetic Resonance Spectroscopy ,Dendritic spine ,Biophysics ,IDP ,Nerve Tissue Proteins ,Prodomain ,Hippocampal formation ,Biochemistry ,Article ,Biomaterials ,03 medical and health sciences ,0302 clinical medicine ,Neurotrophic factors ,medicine ,Binding site ,Receptor ,Growth cone ,Binding Sites ,Chemistry ,Brain-Derived Neurotrophic Factor ,Metals and Alloys ,purl.org/becyt/ford/3.1 [https] ,Human brain ,Cell biology ,Zinc ,BDNF ,030104 developmental biology ,medicine.anatomical_structure ,Chemistry (miscellaneous) ,purl.org/becyt/ford/3 [https] ,030217 neurology & neurosurgery ,Protein Binding - Abstract
Human brain derived neurotrophic factor (BDNF) encodes a protein product consisting of a C-terminal mature domain (mature BDNF) and an N-terminal prodomain, which is an intrinsically disordered protein. A common single nucleotide polymorphism in humans results in a methionine substitution for valine at position 66 of the prodomain, and is associated with memory deficits, depression and anxiety disorders. The BDNF Met66 prodomain, but not the Val66 prodomain, promotes rapid structural remodeling of hippocampal neurons’ growth cones and dendritic spines by interacting directly with the SorCS2 receptor. While it has been reported that the Met66 and Val66 prodomains exhibit only modest differences in structural propensities in the apo state, here we show that Val66 and Met66 prodomains differentially bind zinc (Zn). Zn2+ binds with higher affinity and more broadly impacts residues on the Met66 prodomain compared to the Val66 prodomain as shown by NMR and ITC. Zn2+ binding to the Met66 and Val66 prodomains results in distinct conformational and macroscopic differences observed by NMR, light scattering and cryoEM. To determine if Zn2+ mediated conformational change in the Met66 prodomain is required for biological effect, we mutated His40, a Zn2+ binding site, and observed a loss of Met66 prodomain bioactivity. As the His40 site is distant from the known region of the prodomain involved in receptor binding, we suggest that Met66 prodomain bioactivity involves His40 mediated stabilization of the multimeric structure. Our results point to the necessity of a Zn2+-mediated higher order molecular assembly of the Met66 prodomain to mediate neuronal remodeling. Fil: Wang, Jing. Weill Cornell Medicine; Estados Unidos Fil: Anastasia Gonzalez, Agustin. Weill Cornell Medicine; Estados Unidos. Consejo Nacional de Investigaciones Científicas y Técnicas. Centro Científico Tecnológico Conicet - Córdoba. Instituto de Investigación Médica Mercedes y Martín Ferreyra. Universidad Nacional de Córdoba. Instituto de Investigación Médica Mercedes y Martín Ferreyra; Argentina Fil: Bains, Henrietta. Weill Cornell Medicine; Estados Unidos Fil: Giza, Joanna I.. Borough of Manhattan Community College; Estados Unidos Fil: Clossey, David G.. Weill Cornell Medicine; Estados Unidos Fil: Deng, Jingjing. New York University. School of Medicine; Estados Unidos Fil: Neubert, Thomas A.. New York University Langone Health; Estados Unidos Fil: Rice, William J.. New York University Langone Health; Estados Unidos Fil: Lee, Francis S.. Weill Cornell Medicine; Estados Unidos Fil: Hempstead, Barbara L.. Weill Cornell Medicine; Estados Unidos Fil: Bracken, Clay. Weill Cornell Medicine; Estados Unidos
- Published
- 2020
- Full Text
- View/download PDF
16. SorCS2 is required for social memory and trafficking of the NMDA receptor
- Author
-
Iva Dincheva, Teresa A. Milner, Tina Marinic, Joanna Giza, Francis S. Lee, Jianmin Yang, Barbara L. Hempstead, Deqiang Jing, Anjali M. Rajadhyaksha, and Qian Ma
- Subjects
0301 basic medicine ,Dendritic spine ,Olfaction ,AMPA receptor ,Hippocampal formation ,Biology ,medicine.disease ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Psychiatry and Mental health ,030104 developmental biology ,0302 clinical medicine ,nervous system ,Schizophrenia ,medicine ,NMDA receptor ,Receptor ,Molecular Biology ,Neuroscience ,Postsynaptic density ,030217 neurology & neurosurgery - Abstract
Social memory processing requires functional CA2 neurons, however the specific mechanisms that regulate their activity are poorly understood. Here, we document that SorCS2, a member of the family of the Vps10 family of sorting receptors, is highly expressed in pyramidal neurons of CA2, as well as ventral CA1, a circuit implicated in social memory. SorCS2 specifically localizes to the postsynaptic density and endosomes within dendritic spines of CA2 neurons. We have discovered that SorCS2 is a selective regulator of NMDA receptor surface trafficking in hippocampal neurons, without altering AMPA receptor trafficking. In addition, SorCS2 regulates dendritic spine density in CA2 neurons where SorCS2 expression is enriched, but not in dorsal CA1 neurons, which normally express very low levels of this protein. To specifically test the role of SorCS2 in behavior, we generated a novel SorCS2-deficient mouse, and identify a significant social memory deficit, with no change in sociability, olfaction, anxiety, or several hippocampal-dependent behaviors. Mutations in sorCS2 have been associated with bipolar disease, schizophrenia, and attention deficient-hyperactivity disorder, and abnormalities in social memory are core components of these neuropsychiatric conditions. Thus, our findings provide a new mechanism for social memory formation, through regulating synaptic receptor trafficking in pyramidal neurons by SorCS2.
- Published
- 2020
- Full Text
- View/download PDF
17. Corticosterone induces discrete epigenetic signatures in the dorsal and ventral hippocampus that depend upon sex and genotype: focus on methylated Nr3c1 gene
- Author
-
Salvatore G. Caradonna, Nathan R. Einhorn, Vikram Saudagar, Huzefa Khalil, Gordon H. Petty, Axel Lihagen, Claire LeFloch, Francis S. Lee, Huda Akil, Alessandro Guidotti, Bruce S. McEwen, Eleonora Gatta, and Jordan Marrocco
- Subjects
Male ,Hypothalamo-Hypophyseal System ,Genotype ,Pituitary-Adrenal System ,Hippocampus ,Epigenesis, Genetic ,Cellular and Molecular Neuroscience ,Psychiatry and Mental health ,Mice ,Receptors, Glucocorticoid ,Animals ,Female ,Corticosterone ,Biological Psychiatry - Abstract
The genomic effects of circulating glucocorticoids are particularly relevant in cortico-limbic structures, which express a high concentration of steroid hormone receptors. To date, no studies have investigated genomic differences in hippocampal subregions, namely the dorsal (dHPC) and ventral (vHPC) hippocampus, in preclinical models treated with exogenous glucocorticoids. Chronic oral corticosterone (CORT) in mouse is a pharmacological approach that disrupts the activity of the hypothalamic-pituitary-adrenal axis, increases affective behavior, and induces genomic changes after stress in the HPC of wildtype (WT) mice and mice heterozygous for the gene coding for brain-derived neurotrophic factor Val66Met (hMet), a variant associated with genetic susceptibility to stress. Using RNA-sequencing, we investigated the genomic signatures of oral CORT in the dHPC and vHPC of WT and hMet male and female mice, and examined sex and genotype differences in response to oral CORT. Males under CORT showed lower glycemia and increased anxiety- and depression-like behavior compared to females that showed instead opposite affective behavior in response to CORT. Rank–rank-hypergeometric overlap (RRHO) was used to identify genes from a continuous gradient of significancy that were concordant across groups. RRHO showed that CORT-induced differentially expressed genes (DEGs) in WT mice and hMet mice converged in the dHPC of males and females, while in the vHPC, DEGs converged in males and diverged in females. The vHPC showed a higher number of DEGs compared to the dHPC and exhibited sex differences related to glucocorticoid receptor (GR)-binding genes and epigenetic modifiers. Methyl-DNA-immunoprecipitation in the vHPC revealed differential methylation of the exons 1C and 1F of the GR gene (Nr3c1) in hMet females. Together, we report behavioral and endocrinological sex differences in response to CORT, as well as epigenetic signatures that i) differ in the dHPC and vHPC,ii) are distinct in males and females, and iii) implicate differential methylation of Nr3c1 selectively in hMet females.
- Published
- 2022
18. Dataset related to the article: 'The α2-adrenergic receptor pathway modulating depression influences the risk of arterial thrombosis associated with BDNFVal66Met polymorphism'
- Author
-
Leonardo Sandrini, Patrizia Amadio, Alessandro Ieraci, Alessandro Malara, Josè Pablo Werba, Paolo Maria Soprano, Alessandra Balduini, Marta Zarà, Alice Bonomi, Fabrizio Veglia, Gualtiero I Colombo, Maurizio Popoli, Francis S. Lee, Elena Tremoli, and Silvia Stella Barbieri
- Abstract
This record contains raw data related to the article:"The α2-adrenergic receptor pathway modulating depression influences the risk of arterial thrombosis associated with BDNFVal66Met polymorphism" Abstract Depression is associated with thrombotic risk and arterial events, its proper management is strongly recommended incoronary artery disease(CAD) patients. We have previously shown that the Brain-Derived Neurotrophic Factor (BDNF)Val66Met polymorphism, related to depression, is associated witharterial thrombosisin mice, and with an increased risk ofacute myocardial infarctionin humans. Herein, expanding the previous findings on BDNFVal66Met polymorphism, we show thatdesipramine, anorepinephrinereuptake-inhibitor, rescues behavioral impairments, reduces the arterial thrombosis risk, abolishes pathological coagulation and platelet hyper-reactivity, normalizes leukocyte, platelet, and bone marrowmegakaryocytenumber and restores physiological norepinephrine levels in homozygous knock-in BDNF Val66Met (BDNFMet/Met) mice. Thein vitrodata confirm the enhancedprocoagulant activityand the alpha2A-adrenergic receptor (α2A-ADR) overexpression found in BDNFMet/Metmice and we provide evidence that, in presence of Met variant, norepinephrine is crucial to up-regulate procoagulant activity and to enhance platelet generation. The α2-ADR antagonistrauwolscinerescues the prothrombotic phenotype in BDNFMet/Metmice and reduces procoagulant activity and platelet generation in cells transfected with BDNFMetplasmid or exposed to pro-BDNFMetpeptide. Finally, we show that homozygous BDNFMet/MetCAD patients have hyper-reactive platelets overexpressing abundant α2A-ADR. The great proplatelet release from their megakaryocytes well reflects their higher circulating platelet number compared to BDNFVal/Valpatients. These data reveal an unprecedented described role of Met allele in the dysregulation of norepinephrine/α2A-ADR pathway that may explain the predisposition to arterial thrombosis. Overall, the development of α2A-ADR inhibitors might represent apharmacological treatmentfor depression-associated thrombotic conditions in this specific subgroup of CAD patients.
- Published
- 2022
- Full Text
- View/download PDF
19. Human immunomodulatory ligand B7-1 mediates synaptic remodeling via the p75 neurotrophin receptor
- Author
-
Nicholas C. Morano, Roshelle S. Smith, Victor Danelon, Ryan Schreiner, Uttsav Patel, Natalia G. Herrera, Carla Smith, Steven M. Olson, Michelle K. Laerke, Alev Celikgil, Scott J. Garforth, Sarah C. Garrett-Thomson, Francis S. Lee, Barbara L. Hempstead, and Steven C. Almo
- Subjects
Neurons ,Mice ,Synapses ,B7-1 Antigen ,Animals ,Humans ,CTLA-4 Antigen ,General Medicine ,Receptors, Nerve Growth Factor ,Receptor, Nerve Growth Factor ,Rats - Abstract
Cell surface receptors, ligands, and adhesion molecules underlie development, circuit formation, and synaptic function of the central nervous system and represent important therapeutic targets for many neuropathologies. The functional contributions of interactions between cell surface proteins of neurons and nonneuronal cells have not been fully addressed. Using an unbiased protein-protein interaction screen, we showed that the human immunomodulatory ligand B7-1 (hB7-1) interacts with the p75 neurotrophin receptor (p75NTR) and that the B7-1:p75NTR interaction is a recent evolutionary adaptation present in humans and other primates, but absent in mice, rats, and other lower mammals. The surface of hB7-1 that engages p75NTR overlaps with the hB7-1 surface involved in CTLA-4/CD28 recognition, and these molecules directly compete for binding to p75NTR. Soluble or membrane-bound hB7-1 altered dendritic morphology of cultured hippocampal neurons, with loss of the postsynaptic protein PSD95 in a p75NTR-dependent manner. Abatacept, an FDA-approved therapeutic (CTLA-4-hFc fusion) inhibited these processes. In vivo injection of hB7-1 into the murine subiculum, a hippocampal region affected in Alzheimer's disease, resulted in p75NTR-dependent pruning of dendritic spines. Here, we report the biochemical interaction between B7-1 and p75NTR, describe biological effects on neuronal morphology, and identify a therapeutic opportunity for treatment of neuroinflammatory diseases.
- Published
- 2021
20. The α
- Author
-
Leonardo, Sandrini, Patrizia, Amadio, Alessandro, Ieraci, Alessandro, Malara, José P, Werba, Paolo M, Soprano, Alessandra, Balduini, Marta, Zarà, Alice, Bonomi, Fabrizio, Veglia, Gualtiero I, Colombo, Maurizio, Popoli, Francis S, Lee, Elena, Tremoli, and Silvia S, Barbieri
- Subjects
Aged, 80 and over ,Male ,Depression ,Brain-Derived Neurotrophic Factor ,Desipramine ,Thrombosis ,Coronary Artery Disease ,Middle Aged ,Polymorphism, Single Nucleotide ,Mice ,Norepinephrine ,Receptors, Adrenergic, alpha-2 ,Animals ,Humans ,Female ,Blood Coagulation ,Aged - Abstract
Depression is associated with thrombotic risk and arterial events, its proper management is strongly recommended in coronary artery disease (CAD) patients. We have previously shown that the Brain-Derived Neurotrophic Factor (BDNF)Val66Met polymorphism, related to depression, is associated with arterial thrombosis in mice, and with an increased risk of acute myocardial infarction in humans. Herein, expanding the previous findings on BDNFVal66Met polymorphism, we show that desipramine, a norepinephrine reuptake-inhibitor, rescues behavioral impairments, reduces the arterial thrombosis risk, abolishes pathological coagulation and platelet hyper-reactivity, normalizes leukocyte, platelet, and bone marrow megakaryocyte number and restores physiological norepinephrine levels in homozygous knock-in BDNF Val66Met (BDNF
- Published
- 2021
21. The role of BDNF in mediating the prophylactic effects of (R,S)-ketamine on fear generalization and extinction
- Author
-
James D. Ryan, Nathaniel Tse, Chienchun Huang, Ruirong Yang, and Francis S. Lee
- Subjects
Cellular and Molecular Neuroscience ,Psychiatry and Mental health ,Mice ,Brain-Derived Neurotrophic Factor ,Animals ,Ketamine ,Fear ,Hippocampus ,Biological Psychiatry ,Generalization, Psychological - Abstract
Fear generalization is a conserved survival mechanism that can become maladaptive in the face of traumatic situations, a feature central to certain anxiety disorders including posttraumatic stress disorder (PTSD). However, the neural circuitry and molecular mechanisms underlying fear generalization remain unclear. Recent studies have shown that prophylactic treatment with (R,S)-ketamine confers protective effects in stress-induced depressive behaviors and enhances contextual fear discrimination, but the extent to which these effects extend to fear generalization after auditory fear conditioning remains unclear. Here, we build on this work by using a behavioral model of fear generalization in mice involving foot shocks with differential intensity levels during auditory fear conditioning. We find that prophylactic (R,S)-ketamine treatment exerts protective effects that results in enhanced fear discrimination in wild type mice. As the growth factor, brain-derived neurotrophic factor (BDNF), has been shown to mediate the rapid antidepressant actions of (R,S)-ketamine, we used a loss-of-function BDNF mouse line (BDNF Val66Met) to determine whether BDNF is involved in (R,S)-ketamine’s prophylactic effects on fear generalization. We found that BDNF Val66Met mice were resistant to the protective effects of prophylactic (R,S)-ketamine administration on fear generalization and extinction. We then used fiber photometry to parse out underlying neural activity and found that in the ventral hippocampus there were significant fear generalization-dependent patterns of activity for wild type and BDNF Val66Met mice that were altered by prophylactic (R,S)-ketamine treatment. Overall, these findings indicate a role for the ventral hippocampus and BDNF signaling in modulating the mitigating effects of prophylactic (R,S)-ketamine treatment on generalized fear.
- Published
- 2021
22. Genomic Modules and Intramodular Network Convergency of Susceptibility and Resilience in Multimodeled Stress in Male Mice
- Author
-
Huda Akil, Einhorn Nr, Francis S. Lee, Bruce S. McEwen, Caradonna Sg, Huzefa Khalil, Shen M, Michael J. Meaney, Jordan Marrocco, Tie-Yuan Zhang, Parent C, O’Toole N, and Xianglan Wen
- Subjects
Genetics ,Social defeat ,Environmental enrichment ,education.field_of_study ,Neurotrophic factors ,Population ,Wild type ,Gene regulatory network ,Genetic predisposition ,Biology ,education ,Gene - Abstract
The multifactorial etiology of stress-related disorders is a challenge in developing synchronized medical standards for treatment and diagnosis. It is largely unknown whether there exists molecular convergence in preclinical models of stress generated using disparate construct validity. Using RNA-sequencing (RNA-seq), we investigated the genomic signatures in the ventral hippocampus, which mostly regulates affective behavior, in mouse models that recapitulate the hallmarks of anxiety and depression. Chronic oral corticosterone (CORT), a model that causes a blunted endocrine response to stress, induced anxiety- and depression-like behavior in wildtype mice and mice heterozygous for the gene coding for brain-derived neurotrophic factor (BDNF) Val66Met, a variant associated with genetic susceptibility to stress. In a separate set of mice, chronic social defeat stress led to a susceptible or a resilient population, whose proportion was dependent on housing conditions, standard housing or enriched environment. A rank-rank-hypergeometric (RRHO) analysis of the RNA-seq data set across models demonstrated that in mice treated with CORT and susceptible mice raised in standard housing differentially expressed genes (DEGs) converged toward gene networks involved in similar biological functions. Weighted gene co-expression analysis generated 54 unique modules of interconnected gene hubs, two of which included a combination of all experimental groups and were significantly enriched in DEGs, whose function was consistent with that predicted in the RRHO GO analysis. This multimodel approach showed transcriptional synchrony between models of stress with hormonal, environmental or genetic construct validity shedding light on common genomic drivers that embody the multifaceted nature of stress-related disorders.
- Published
- 2021
- Full Text
- View/download PDF
23. The microbiota regulate neuronal function and fear extinction learning
- Author
-
Sangeeta S. Chavan, Coco Chu, Aviv Regev, Conor Liston, David Artis, Lei Zhou, Frank C. Schroeder, Meghan E. Addorisio, Saya Moriyama, Anfei Li, Tae Hyung Won, Ruirong Yang, Mitchell H. Murdock, Heidi C. Meyer, Fei Teng, Gregory G. Putzel, Adam M. Kressel, Kevin J. Tracey, Nicholas J. Bessman, Deqiang Jing, Hattie Chung, Christopher N. Parkhurst, Tea Tsaava, and Francis S. Lee
- Subjects
Male ,0301 basic medicine ,Cell type ,Dendritic spine ,Dendritic Spines ,Prefrontal Cortex ,Biology ,Article ,Extinction, Psychological ,Feces ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Postsynaptic potential ,Neuroplasticity ,medicine ,Animals ,Germ-Free Life ,Metabolomics ,Premovement neuronal activity ,Autistic Disorder ,Prefrontal cortex ,Cerebrospinal Fluid ,Neurons ,Mice, Inbred BALB C ,Multidisciplinary ,Phenylpropionates ,Microbiota ,Neural Inhibition ,Vagus Nerve ,Fear ,Extinction (psychology) ,Anti-Bacterial Agents ,Mice, Inbred C57BL ,Blood ,030104 developmental biology ,medicine.anatomical_structure ,Schizophrenia ,Neuroglia ,Calcium ,Cues ,Transcriptome ,Indican ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Multicellular organisms have co-evolved with complex consortia of viruses, bacteria, fungi and parasites, collectively referred to as the microbiota1. In mammals, changes in the composition of the microbiota can influence many physiologic processes (including development, metabolism and immune cell function) and are associated with susceptibility to multiple diseases2. Alterations in the microbiota can also modulate host behaviours—such as social activity, stress, and anxiety-related responses—that are linked to diverse neuropsychiatric disorders3. However, the mechanisms by which the microbiota influence neuronal activity and host behaviour remain poorly defined. Here we show that manipulation of the microbiota in antibiotic-treated or germ-free adult mice results in significant deficits in fear extinction learning. Single-nucleus RNA sequencing of the medial prefrontal cortex of the brain revealed significant alterations in gene expression in excitatory neurons, glia and other cell types. Transcranial two-photon imaging showed that deficits in extinction learning after manipulation of the microbiota in adult mice were associated with defective learning-related remodelling of postsynaptic dendritic spines and reduced activity in cue-encoding neurons in the medial prefrontal cortex. In addition, selective re-establishment of the microbiota revealed a limited neonatal developmental window in which microbiota-derived signals can restore normal extinction learning in adulthood. Finally, unbiased metabolomic analysis identified four metabolites that were significantly downregulated in germ-free mice and have been reported to be related to neuropsychiatric disorders in humans and mouse models, suggesting that microbiota-derived compounds may directly affect brain function and behaviour. Together, these data indicate that fear extinction learning requires microbiota-derived signals both during early postnatal neurodevelopment and in adult mice, with implications for our understanding of how diet, infection, and lifestyle influence brain health and subsequent susceptibility to neuropsychiatric disorders. A diverse intestinal microbiota is required for mice to undergo extinction-related neuronal plasticity and normal fear extinction learning.
- Published
- 2019
- Full Text
- View/download PDF
24. Role of BDNF in the development of an OFC-amygdala circuit regulating sociability in mouse and human
- Author
-
Baila S. Hall, Anfei Li, Danielle V. Dellarco, Chienchun Huang, B. J. Casey, Francis S. Lee, Conor Liston, Ruirong Yang, Deqiang Jing, and Ross T Heilberg
- Subjects
0301 basic medicine ,Adolescent ,Biology ,Social identity approach ,Polymorphism, Single Nucleotide ,Amygdala ,Article ,Mice ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Functional neuroimaging ,medicine ,Animals ,Humans ,SNP ,Molecular Biology ,Loss function ,Extramural ,Brain-Derived Neurotrophic Factor ,Social anxiety ,Fear ,Psychiatry and Mental health ,030104 developmental biology ,medicine.anatomical_structure ,Neuroscience ,030217 neurology & neurosurgery ,Social behavior - Abstract
Social deficits are common in many psychiatric disorders. However, due to inadequate tools for manipulating circuit activity in humans and unspecific paradigms for modeling social behaviors in rodents, our understanding of the molecular and circuit mechanisms mediating social behaviors remains relatively limited. Using human functional neuroimaging and rodent fiber photometry, we identified a mOFC-BLA projection that modulates social approach behavior and influences susceptibility to social anxiety. In humans and knock-in mice with a loss of function BDNF SNP (Val66Met), the functionality of this circuit was altered, resulting in social behavioral changes in human and mice. We further showed that the development of this circuit is disrupted in BDNF Met carriers due to insufficient BDNF bioavailability, specifically during a peri-adolescent timeframe. These findings define one mechanism by which social anxiety may stem from altered maturation of orbitofronto-amygdala projections and identify a developmental window in which BDNF-based interventions may have therapeutic potential.
- Published
- 2019
- Full Text
- View/download PDF
25. Translating Developmental Neuroscience to Understand Risk for Psychiatric Disorders
- Author
-
Heidi C. Meyer and Francis S. Lee
- Subjects
medicine.medical_specialty ,Adolescent ,Vulnerability ,Developmental cognitive neuroscience ,Translational research ,03 medical and health sciences ,Child Development ,0302 clinical medicine ,Neuroimaging ,Risk Factors ,Neuroplasticity ,medicine ,Humans ,Child ,Psychiatry ,Psychopathology ,Mental Disorders ,Brain ,Cognition ,Adolescent Development ,medicine.disease ,030227 psychiatry ,Psychiatry and Mental health ,Neurodevelopmental Disorders ,Schizophrenia ,Psychology ,Stress, Psychological ,030217 neurology & neurosurgery - Abstract
The transition from childhood to adulthood represents the developmental time frame in which the majority of psychiatric disorders emerge. Recent efforts to identify risk factors mediating the susceptibility to psychopathology have led to a heightened focus on both typical and atypical trajectories of neural circuit maturation. Mounting evidence has highlighted the immense neural plasticity apparent in the developing brain. Although in many cases adaptive, the capacity for neural circuit alteration also induces a state of vulnerability to environmental perturbations, such that early-life experiences have long-lasting implications for cognitive and emotional functioning in adulthood. The authors outline preclinical and neuroimaging studies of normative human brain circuit development, as well as parallel efforts covered in this issue of the Journal, to identify brain circuit alterations in psychiatric disorders that frequently emerge in developing populations. Continued translational research into the interactive effects of neurobiological development and external factors will be crucial for identifying early-life risk factors that may contribute to the emergence of psychiatric illness and provide the key to optimizing treatments.
- Published
- 2019
- Full Text
- View/download PDF
26. Scn2a severe hypomorphic mutation decreases excitatory synaptic input and causes autism-associated behaviors
- Author
-
Geoffrey S. Pitt, Charlotte C. Bavley, Yared Bayleyen, Rebecca M. Jones, Anfei Li, Anjali M. Rajadhyaksha, Francis S. Lee, Hong-Gang Wang, and Jonathan E. Hackett
- Subjects
Autism Spectrum Disorder ,Mutant ,Mouse models ,Inhibitory postsynaptic potential ,Mice ,Loss of Function Mutation ,Animals ,Correlation of Data ,Cells, Cultured ,Ion channel ,Neurons ,NAV1.2 Voltage-Gated Sodium Channel ,Behavior, Animal ,Chemistry ,Brain ,General Medicine ,Cell biology ,Animal Communication ,Disease Models, Animal ,Electrophysiology ,Gene Expression Regulation ,Ion channels ,Forebrain ,Excitatory postsynaptic potential ,Heterologous expression ,Intracellular ,Research Article ,Neuroscience - Abstract
SCN2A, encoding the neuronal voltage-gated Na+ channel NaV1.2, is one of the most commonly affected loci linked to autism spectrum disorders (ASDs). Most ASD-associated mutations in SCN2A are loss-of-function mutations, but studies examining how such mutations affect neuronal function and whether Scn2a mutant mice display ASD endophenotypes have been inconsistent. We generated a protein truncation variant Scn2a mouse model (Scn2aΔ1898/+) by CRISPR that eliminates the NaV1.2 channel’s distal intracellular C-terminal domain, and we analyzed the molecular and cellular consequences of this variant in a heterologous expression system, in neuronal culture, in brain slices, and in vivo. We also analyzed multiple behaviors in WT and Scn2aΔ1898/+ mice and correlated behaviors with clinical data obtained in human subjects with SCN2A variants. Expression of the NaV1.2 mutant in a heterologous expression system revealed decreased NaV1.2 channel function, and cultured pyramidal neurons isolated from Scn2aΔ1898/+ forebrain showed correspondingly reduced voltage-gated Na+ channel currents without compensation from other CNS voltage-gated Na+ channels. Na+ currents in inhibitory neurons were unaffected. Consistent with loss of voltage-gated Na+ channel currents, Scn2aΔ1898/+ pyramidal neurons displayed reduced excitability in forebrain neuronal culture and reduced excitatory synaptic input onto the pyramidal neurons in brain slices. Scn2aΔ1898/+ mice displayed several behavioral abnormalities, including abnormal social interactions that reflect behavior observed in humans with ASD and with harboring loss-of-function SCN2A variants. This model and its cellular electrophysiological characterizations provide a framework for tracing how a SCN2A loss-of-function variant leads to cellular defects that result in ASD-associated behaviors.
- Published
- 2021
- Full Text
- View/download PDF
27. SLITRK5 is a negative regulator of hedgehog signaling in osteoblasts
- Author
-
Sarfaraz Lalani, Dong Yeon Shin, Seoyeon Bok, Michelle Cung, Abdul G. Khan, Zan Li, Ren Xu, Matthew B. Greenblatt, Jae-Hyuck Shim, Jun Sun, Alisha R. Yallowitz, Na Li, Sofia Jenia Marquez, Ivo C. Lorenz, Tommy E. White, Shawon Debnath, Francis S. Lee, and Mark Eiseman
- Subjects
0301 basic medicine ,animal structures ,Cellular differentiation ,Science ,education ,General Physics and Astronomy ,Nerve Tissue Proteins ,General Biochemistry, Genetics and Molecular Biology ,Article ,Morphogen signalling ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Osteogenesis ,medicine ,Animals ,Humans ,Hedgehog Proteins ,Cilia ,Hedgehog ,Cells, Cultured ,Mice, Knockout ,Multidisciplinary ,Osteoblasts ,Chemistry ,Cilium ,Membrane Proteins ,Bone development ,Osteoblast ,Cell Differentiation ,General Chemistry ,Transmembrane protein ,Hedgehog signaling pathway ,Cell biology ,Patched-1 Receptor ,030104 developmental biology ,medicine.anatomical_structure ,PTCH1 ,embryonic structures ,Signal transduction ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Hedgehog signaling is essential for bone formation, including functioning as a means for the growth plate to drive skeletal mineralization. However, the mechanisms regulating hedgehog signaling specifically in bone-forming osteoblasts are largely unknown. Here, we identified SLIT and NTRK-like protein-5(Slitrk5), a transmembrane protein with few identified functions, as a negative regulator of hedgehog signaling in osteoblasts. Slitrk5 is selectively expressed in osteoblasts and loss of Slitrk5 enhanced osteoblast differentiation in vitro and in vivo. Loss of SLITRK5 in vitro leads to increased hedgehog signaling and overexpression of SLITRK5 in osteoblasts inhibits the induction of targets downstream of hedgehog signaling. Mechanistically, SLITRK5 binds to hedgehog ligands via its extracellular domain and interacts with PTCH1 via its intracellular domain. SLITRK5 is present in the primary cilium, and loss of SLITRK5 enhances SMO ciliary enrichment upon SHH stimulation. Thus, SLITRK5 is a negative regulator of hedgehog signaling in osteoblasts that may be attractive as a therapeutic target to enhance bone formation., Hedgehog signaling is essential for bone formation. Here, the authors show that the transmembrane protein SLITRK5 is a negative regulator of hedgehog signaling in osteoblasts, suggesting it may be a potential therapeutic target to enhance bone formation.
- Published
- 2020
28. Pre-adolescent stress disrupts adult, but not adolescent, safety learning
- Author
-
Danielle M. Gerhard, Francis S. Lee, Paia A. Amelio, and Heidi C. Meyer
- Subjects
Male ,Conditioned inhibition ,Conditioning, Classical ,Pre adolescents ,Anxiety ,Article ,Discrimination Learning ,03 medical and health sciences ,Behavioral Neuroscience ,Mice ,0302 clinical medicine ,Pregnancy ,Stress (linguistics) ,Medicine ,Animals ,Postnatal day ,030304 developmental biology ,0303 health sciences ,Behavior, Animal ,business.industry ,Age Factors ,Fear ,Mice, Inbred C57BL ,Disease Models, Animal ,Inhibition, Psychological ,Conditioning ,Female ,medicine.symptom ,Safety ,business ,030217 neurology & neurosurgery ,Stress, Psychological ,Clinical psychology - Abstract
Anxiety disorders are highly prevalent across the lifespan, although diagnoses peak early in adolescence. As a method for inhibiting fear, safety signals have the potential to augment conventional treatments for anxiety. However, the ability to acquire and use safety signals during adolescence remains unclear. Moreover, the impact of stress on safety learning has received surprisingly little attention given that stress is a major factor preceding anxiety onset. In this study, mice were trained in a discriminative conditioning protocol to facilitate safety learning and were tested for fear inhibition using a conditioned safety signal. Next, independent groups of mice were exposed to chronic unpredictable stress (CUS) conditions between postnatal day 22 and 28, followed by tests for anxiety-like phenotypes or fear inhibition using a safety signal, performed either 24 hours or five weeks following CUS. Pre-adolescent CUS reduced weight in adolescence and this effect endured into adulthood. CUS also increased specific anxiety-like behaviors in adolescence that were unique from the increase in anxiety observed in adulthood. Despite increased anxiety-like behaviors, adolescents were able to learn about and effectively use safety signals to inhibit fear. In contrast, adults that experienced CUS showed a subtle increase in anxiety but had impaired safety signal learning and usage. Together, these findings indicate that pre-adolescent stress has immediate and enduring effects on anxiety-like behaviors but impairs the capacity for conditioned inhibition only following incubation.
- Published
- 2020
29. Impact of
- Author
-
Leonardo, Sandrini, Laura, Castiglioni, Patrizia, Amadio, José Pablo, Werba, Sonia, Eligini, Susanna, Fiorelli, Marta, Zarà, Silvia, Castiglioni, Stefano, Bellosta, Francis S, Lee, Luigi, Sironi, Elena, Tremoli, and Silvia Stella, Barbieri
- Subjects
Aged, 80 and over ,Male ,Mice, Knockout ,BDNF Val66Met polymorphism ,Genotype ,Ventricular Remodeling ,Brain-Derived Neurotrophic Factor ,Macrophages ,macrophage phenotype ,Myocardial Infarction ,Brain ,Middle Aged ,Hippocampus ,Polymorphism, Single Nucleotide ,Article ,Mice ,Phenotype ,myocardial infarction ,cardiovascular disease ,Animals ,Humans ,Aged - Abstract
Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin growth factor family, well known for its role in the homeostasis of the cardiovascular system. Recently, the human BDNF Val66Met single nucleotide polymorphism has been associated with the increased propensity for arterial thrombosis related to acute myocardial infarction (AMI). Using cardiac magnetic resonance imaging and immunohistochemistry analyses, we showed that homozygous mice carrying the human BDNF Val66Met polymorphism (BDNFMet/Met) undergoing left anterior descending (LAD) coronary artery ligation display an adverse cardiac remodeling compared to wild-type (BDNFVal/Val). Interestingly, we observed a persistent presence of pro-inflammatory M1-like macrophages and a reduced accumulation of reparative-like phenotype macrophages (M2-like) in the infarcted heart of mutant mice. Further qPCR analyses showed that BDNFMet/Met peritoneal macrophages are more pro-inflammatory and have a higher migratory ability compared to BDNFVal/Val ones. Finally, macrophages differentiated from circulating monocytes isolated from BDNFMet/Met patients with coronary heart disease displayed the same pro-inflammatory characteristics of the murine ones. In conclusion, the BDNF Val66Met polymorphism predisposes to adverse cardiac remodeling after myocardial infarction in a mouse model and affects macrophage phenotype in both humans and mice. These results provide a new cellular mechanism by which this human BDNF genetic variant could influence cardiovascular disease.
- Published
- 2020
30. Endocannabinoid genetic variation enhances vulnerability to THC reward in adolescent female mice
- Author
-
Ruirong Yang, Chienchun Huang, Francis S. Lee, Deqiang Jing, Anjali M. Rajadhyaksha, Virginia M. Pickel, Caitlin E. Burgdorf, Teresa A. Milner, Matthew N. Hill, and Ken Mackie
- Subjects
Male ,Aging ,Tyrosine 3-Monooxygenase ,medicine.medical_treatment ,Biology ,Nucleus accumbens ,Inhibitory postsynaptic potential ,Choice Behavior ,Polymorphism, Single Nucleotide ,Nucleus Accumbens ,Amidohydrolases ,03 medical and health sciences ,0302 clinical medicine ,Receptor, Cannabinoid, CB1 ,Reward ,mental disorders ,Genetic variation ,medicine ,Animals ,Dronabinol ,Cannabis Dependence ,Research Articles ,Multidisciplinary ,musculoskeletal, neural, and ocular physiology ,organic chemicals ,Ventral Tegmental Area ,SciAdv r-articles ,Genetic Variation ,Endocannabinoid system ,Axons ,030227 psychiatry ,Mice, Inbred C57BL ,Ventral tegmental area ,medicine.anatomical_structure ,nervous system ,Excitatory postsynaptic potential ,lipids (amino acids, peptides, and proteins) ,Female ,Cannabinoid ,Nerve Net ,Neuroscience ,psychological phenomena and processes ,030217 neurology & neurosurgery ,Research Article ,Endocannabinoids - Abstract
A genetic variant in the endocannabinoid system enhances vulnerability to THC reward in adolescent females., Adolescence represents a developmental period with the highest risk for initiating cannabis use. Little is known about whether genetic variation in the endocannabinoid system alters mesolimbic reward circuitry to produce vulnerability to the rewarding properties of the exogenous cannabinoid Δ9-tetrahydrocannabinol (THC). Using a genetic knock-in mouse model (FAAHC/A) that biologically recapitulates the human polymorphism associated with problematic drug use, we find that in adolescent female mice, but not male mice, this FAAH polymorphism enhances the mesolimbic dopamine circuitry projecting from the ventral tegmental area (VTA) to the nucleus accumbens (NAc) and alters cannabinoid receptor 1 (CB1R) levels at inhibitory and excitatory terminals in the VTA. These developmental changes collectively increase vulnerability of adolescent female FAAHC/A mice to THC preference that persists into adulthood. Together, these findings suggest that this endocannabinoid genetic variant is a contributing factor for increased susceptibility to cannabis dependence in adolescent females.
- Published
- 2020
- Full Text
- View/download PDF
31. TrkB deubiquitination by USP8 regulates receptor levels and BDNF-dependent neuronal differentiation
- Author
-
Rubén Deogracias, Carlos Martín-Rodríguez, Francis S. Lee, Juan Carlos Arevalo, Begoña Anta, Francisco J González-Calvo, Minseok Song, and Deqiang Jing
- Subjects
Tropomyosin receptor kinase B ,Hippocampus ,Receptor tyrosine kinase ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Neurotrophic factors ,Endopeptidases ,medicine ,Humans ,Receptor, trkB ,Receptor ,Cells, Cultured ,030304 developmental biology ,Neurons ,0303 health sciences ,Gene knockdown ,Membrane Glycoproteins ,Endosomal Sorting Complexes Required for Transport ,biology ,Brain-Derived Neurotrophic Factor ,musculoskeletal, neural, and ocular physiology ,Cell Biology ,Cell biology ,medicine.anatomical_structure ,nervous system ,embryonic structures ,biology.protein ,Neuron ,Ubiquitin Thiolesterase ,030217 neurology & neurosurgery ,Research Article ,Signal Transduction ,Neurotrophin - Abstract
Ubiquitylation of receptor tyrosine kinases (RTKs) regulates both the levels and functions of these receptors. The neurotrophin receptor TrkB (also known as NTRK2), a RTK, is ubiquitylated upon activation by brain-derived neurotrophic factor (BDNF) binding. Although TrkB ubiquitylation has been demonstrated, there is a lack of knowledge regarding the precise repertoire of proteins that regulates TrkB ubiquitylation. Here, we provide mechanistic evidence indicating that ubiquitin carboxyl-terminal hydrolase 8 (USP8) modulates BDNF- and TrkB-dependent neuronal differentiation. USP8 binds to the C-terminus of TrkB using its microtubule-interacting domain (MIT). Immunopurified USP8 deubiquitylates TrkB in vitro, whereas knockdown of USP8 results in enhanced ubiquitylation of TrkB upon BDNF treatment in neurons. As a consequence of USP8 depletion, TrkB levels and its activation are reduced. Moreover, USP8 protein regulates the differentiation and correct BDNF-dependent dendritic formation of hippocampal neurons in vitro and in vivo. We conclude that USP8 positively regulates the levels and activation of TrkB, modulating BDNF-dependent neuronal differentiation. This article has an associated First Person interview with the first author of the paper.
- Published
- 2020
- Full Text
- View/download PDF
32. A contributing gene for cannabis dependence
- Author
-
Francis S. Lee, Caitlin E. Burgdorf, and Anjali M. Rajadhyaksha
- Subjects
Genetics ,Biology ,Cannabis Dependence ,Gene - Published
- 2020
- Full Text
- View/download PDF
33. The α2-adrenergic receptor pathway modulating depression influences the risk of arterial thrombosis associated with BDNFVal66Met polymorphism
- Author
-
Leonardo Sandrini, Patrizia Amadio, Alessandro Ieraci, Alessandro Malara, José P. Werba, Paolo M. Soprano, Alessandra Balduini, Marta Zarà, Alice Bonomi, Fabrizio Veglia, Gualtiero I. Colombo, Maurizio Popoli, Francis S. Lee, Elena Tremoli, and Silvia S. Barbieri
- Subjects
Anxiety/depression ,Platelets ,Pharmacology ,BDNF Val66Met polymorphism ,Thrombosis ,Procoagulant activity ,Therapeutics. Pharmacology ,RM1-950 ,General Medicine ,α2-adrenergic receptor - Abstract
Depression is associated with thrombotic risk and arterial events, its proper management is strongly recommended in coronary artery disease (CAD) patients. We have previously shown that the Brain-Derived Neurotrophic Factor (BDNF)Val66Met polymorphism, related to depression, is associated with arterial thrombosis in mice, and with an increased risk of acute myocardial infarction in humans. Herein, expanding the previous findings on BDNFVal66Met polymorphism, we show that desipramine, a norepinephrine reuptake-inhibitor, rescues behavioral impairments, reduces the arterial thrombosis risk, abolishes pathological coagulation and platelet hyper-reactivity, normalizes leukocyte, platelet, and bone marrow megakaryocyte number and restores physiological norepinephrine levels in homozygous knock-in BDNF Val66Met (BDNFMet/Met) mice. The in vitro data confirm the enhanced procoagulant activity and the alpha2A-adrenergic receptor (α2A-ADR) overexpression found in BDNFMet/Met mice and we provide evidence that, in presence of Met variant, norepinephrine is crucial to up-regulate procoagulant activity and to enhance platelet generation. The α2-ADR antagonist rauwolscine rescues the prothrombotic phenotype in BDNFMet/Met mice and reduces procoagulant activity and platelet generation in cells transfected with BDNFMet plasmid or exposed to pro-BDNFMet peptide. Finally, we show that homozygous BDNFMet/Met CAD patients have hyper-reactive platelets overexpressing abundant α2A-ADR. The great proplatelet release from their megakaryocytes well reflects their higher circulating platelet number compared to BDNFVal/Val patients. These data reveal an unprecedented described role of Met allele in the dysregulation of norepinephrine/α2A-ADR pathway that may explain the predisposition to arterial thrombosis. Overall, the development of α2A-ADR inhibitors might represent a pharmacological treatment for depression-associated thrombotic conditions in this specific subgroup of CAD patients.
- Published
- 2022
- Full Text
- View/download PDF
34. Global epigenetic analysis of BDNF Val66Met mice hippocampus reveals changes in dendrite and spine remodeling genes
- Author
-
Stefano Corna, Alessandra Mallei, Maurizio Popoli, Alessandro Ieraci, Francis S. Lee, and Daniela Tardito
- Subjects
Male ,0301 basic medicine ,Cognitive Neuroscience ,Mice, Transgenic ,Hippocampus ,Epigenesis, Genetic ,Histones ,03 medical and health sciences ,Histone H3 ,0302 clinical medicine ,Neurotrophic factors ,Gene expression ,Animals ,Humans ,Gene Knock-In Techniques ,Epigenetics ,beta Catenin ,Brain-derived neurotrophic factor ,Polymorphism, Genetic ,biology ,Brain-Derived Neurotrophic Factor ,Computational Biology ,Dendrites ,Wnt Proteins ,Reelin Protein ,030104 developmental biology ,Histone ,Acetylation ,biology.protein ,Neuroscience ,Chromatin immunoprecipitation ,030217 neurology & neurosurgery - Abstract
Brain-derived neurotrophic factor (BDNF), a neurotrophin highly expressed in the hippocampus, plays crucial roles in cognition, neuroplasticity, synaptic function, and dendritic remodeling. The common human Val66Met polymorphism of BDNF has been implicated in the pathophysiology of neuropsychiatric and neurodegenerative disorders, and in the outcome of pro-adaptive and therapeutic treatments. Altered gene-expression profile has been previously shown in BDNF Val66Met knock-in mice, which recapitulate the phenotypic hallmarks of individuals carrying the BDNF Met allele. The aim of this study was to investigate the impact of the BDNF Val66Met polymorphism in the knock-in mouse model on two hippocampal epigenetic marks for transcriptional repression and activation, respectively: trimethylation of lysine 27 on histone H3 (H3K27me3) and acetylation of histone H3 (AcH3), using a genome-wide approach. Chromatin immunoprecipitation followed by deep sequencing of immunoprecipitated DNA (ChIP-Seq) was carried out with specific antibodies for H3K27me3 and AcH3. Our results revealed broad alteration of H3K27me3 and AcH3 marks association profiles in BDNFMet/Met , compared to BDNFVal/Val mice. Bioinformatics analysis showed changes in several biological functions and related pathways, affected by the presence of the polymorphism. In particular, a number of networks of functional interaction contained BDNF as central node. Quantitative PCR analysis confirmed epigenetically related significant changes in the expression of five genes: Dvl1, Nos3, Reln, Lypd6, and Sh3gl2. The first three are involved in dendrite and spine remodeling, morphological features altered in BDNFMet/Met mice. This work in homozygous knock-in mice shows that the human BDNF Val66Met polymorphism induces an array of histone H3 epigenetic modifications, in turn altering the expression of select genes crucial for structural and functional neuronal features.
- Published
- 2018
- Full Text
- View/download PDF
35. Protective effects of elevated anandamide on stress and fear-related behaviors: translational evidence from humans and mice
- Author
-
Matthew N. Hill, Haley A. Vecchiarelli, Lovisa Holm, Niclas Stensson, Georgia Balsevich, Bijar Ghafouri, Anna Asratian, Daniel Nätt, Gaëlle Augier, Johan Lindé, Primavera A. Spagnolo, Francis S. Lee, Robert J. Aukema, Markus Heilig, and Leah M. Mayo
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Adolescent ,Polyunsaturated Alkamides ,Arachidonic Acids ,Protective Agents ,Amygdala ,Amidohydrolases ,Extinction, Psychological ,Stress Disorders, Post-Traumatic ,Mice ,Young Adult ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,0302 clinical medicine ,Fatty acid amide hydrolase ,Internal medicine ,Genetic model ,Animals ,Humans ,Medicine ,Prefrontal cortex ,Molecular Biology ,Prolonged exposure therapy ,business.industry ,Fear ,Extinction (psychology) ,Anandamide ,Psychiatry and Mental health ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Humanized mouse ,Female ,lipids (amino acids, peptides, and proteins) ,business ,030217 neurology & neurosurgery ,Endocannabinoids - Abstract
Post-traumatic stress disorder (PTSD) is a common, debilitating condition with limited treatment options. Extinction of fear memories through prolonged exposure therapy, the primary evidence-based behavioral treatment for PTSD, has only partial efficacy. In mice, pharmacological inhibition of fatty acid amide hydrolase (FAAH) produces elevated levels of anandamide (AEA) and promotes fear extinction, suggesting that FAAH inhibitors may aid fear extinction-based treatments. A human FAAH 385C->A substitution encodes an FAAH enzyme with reduced catabolic efficacy. Individuals homozygous for the FAAH 385A allele may therefore offer a genetic model to evaluate the impact of elevations in AEA signaling in humans, helping to inform whether FAAH inhibitors have the potential to facilitate fear extinction therapy for PTSD. To overcome the challenge posed by low frequency of the AA genotype (appr. 5%), we prospectively genotyped 423 individuals to examine the balanced groups of CC, AC, and AA individuals (n = 25/group). Consistent with its loss-of-function nature, the A allele was dose dependently associated with elevated basal AEA levels, facilitated fear extinction, and enhanced the extinction recall. Moreover, the A-allele homozygotes were protected against stress-induced decreases in AEA and negative emotional consequences of stress. In a humanized mouse model, AA homozygous mice were similarly protected against stress-induced decreases in AEA, both in the periphery, and also in the amygdala and prefrontal cortex, brain structures critically involved in fear extinction and regulation of stress responses. Collectively, these data suggest that AEA signaling can temper aspects of the stress response and that FAAH inhibition may aid the treatment for stress-related psychiatric disorders, such as PTSD.
- Published
- 2018
- Full Text
- View/download PDF
36. Acetyl- <scp>l</scp> -carnitine deficiency in patients with major depressive disorder
- Author
-
James W. Murrough, Sarah P. Young, Ashly Albright, Francis S. Lee, Natalie L. Rasgon, James Beasley, James H. Kocsis, Benedetta Bigio, Carla Nasca, Bruce S. McEwen, David S. Millington, Aleksander A. Mathé, and Marin Kautz
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Hippocampal formation ,03 medical and health sciences ,Glutamatergic ,Sex Factors ,0302 clinical medicine ,Carnitine ,Commentaries ,Internal medicine ,medicine ,Humans ,Epigenetics ,Depression (differential diagnoses) ,Aged ,Depressive Disorder, Major ,Multidisciplinary ,business.industry ,Age Factors ,Middle Aged ,medicine.disease ,030104 developmental biology ,Endophenotype ,Major depressive disorder ,Biomarker (medicine) ,Female ,Age of onset ,Acetylcarnitine ,business ,030217 neurology & neurosurgery - Abstract
The lack of biomarkers to identify target populations greatly limits the promise of precision medicine for major depressive disorder (MDD), a primary cause of ill health and disability. The endogenously produced molecule acetyl-l-carnitine (LAC) is critical for hippocampal function and several behavioral domains. In rodents with depressive-like traits, LAC levels are markedly decreased and signal abnormal hippocampal glutamatergic function and dendritic plasticity. LAC supplementation induces rapid and lasting antidepressant-like effects via epigenetic mechanisms of histone acetylation. This mechanistic model led us to evaluate LAC levels in humans. We found that LAC levels, and not those of free carnitine, were decreased in patients with MDD compared with age- and sex-matched healthy controls in two independent study centers. Secondary exploratory analyses showed that the degree of LAC deficiency reflected both the severity and age of onset of MDD. Moreover, these analyses showed that the decrease in LAC was larger in patients with a history of treatment-resistant depression (TRD), among whom childhood trauma and, specifically, a history of emotional neglect and being female, predicted the decreased LAC. These findings suggest that LAC may serve as a candidate biomarker to help diagnose a clinical endophenotype of MDD characterized by decreased LAC, greater severity, and earlier onset as well as a history of childhood trauma in patients with TRD. Together with studies in rodents, these translational findings support further exploration of LAC as a therapeutic target that may help to define individualized treatments in biologically based depression subtype consistent with the spirit of precision medicine.
- Published
- 2018
- Full Text
- View/download PDF
37. Role for fatty acid amide hydrolase (FAAH) in the leptin-mediated effects on feeding and energy balance
- Author
-
Zhiying Li, Francis S. Lee, Martin A. Sticht, Georgia Balsevich, Cecilia J. Hillard, Nicole P. Bowles, Bruce S. McEwen, Stephanie L. Borgland, Prasanth K. Chelikani, Matthew N. Hill, Arashdeep Singh, and Tiffany T.-Y. Lee
- Subjects
Leptin ,Male ,0301 basic medicine ,medicine.medical_specialty ,Polyunsaturated Alkamides ,Hypothalamus ,Arachidonic Acids ,Amidohydrolases ,Eating ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Fatty acid amide hydrolase ,Internal medicine ,medicine ,Animals ,Gene Knock-In Techniques ,Mice, Knockout ,2. Zero hunger ,Polymorphism, Genetic ,Multidisciplinary ,Chemistry ,Body Weight ,digestive, oral, and skin physiology ,Metabolism ,Anandamide ,Biological Sciences ,Dietary Fats ,Endocannabinoid system ,030104 developmental biology ,Endocrinology ,nervous system ,lipids (amino acids, peptides, and proteins) ,Ghrelin ,Energy Metabolism ,hormones, hormone substitutes, and hormone antagonists ,psychological phenomena and processes ,Endocannabinoids ,Hormone - Abstract
Endocannabinoid signaling regulates feeding and metabolic processes and has been linked to obesity development. Several hormonal signals, such as glucocorticoids and ghrelin, regulate feeding and metabolism by engaging the endocannabinoid system. Similarly, studies have suggested that leptin interacts with the endocannabinoid system, yet the mechanism and functional relevance of this interaction remain elusive. Therefore, we explored the interaction between leptin and endocannabinoid signaling with a focus on fatty acid amide hydrolase (FAAH), the primary degradative enzyme for the endocannabinoid N-arachidonoylethanolamine (anandamide; AEA). Mice deficient in leptin exhibited elevated hypothalamic AEA levels and reductions in FAAH activity while leptin administration to WT mice reduced AEA content and increased FAAH activity. Following high fat diet exposure, mice developed resistance to the effects of leptin administration on hypothalamic AEA content and FAAH activity. At a functional level, pharmacological inhibition of FAAH was sufficient to prevent leptin-mediated effects on body weight and food intake. Using a novel knock-in mouse model recapitulating a common human polymorphism (FAAH C385A; rs324420), which reduces FAAH activity, we investigated whether human genetic variance in FAAH affects leptin sensitivity. While WT (CC) mice were sensitive to leptin-induced reductions in food intake and body weight gain, low-expressing FAAH (AA) mice were unresponsive. These data demonstrate that FAAH activity is required for leptin's hypophagic effects and, at a translational level, suggest that a genetic variant in the FAAH gene contributes to differences in leptin sensitivity in human populations.
- Published
- 2018
- Full Text
- View/download PDF
38. Quantitative validation of immunofluorescence and lectin staining using reduced CLARITY acrylamide formulations
- Author
-
Vivek Kumar, Stanley J. Watson, Karl Deisseroth, Jack D. Barchas, Ben R. Martin, Richard M. Myers, Alan F. Schatzberg, David M. Krolewski, Francis S. Lee, Raju Tomer, William E. Bunney, and Huda Akil
- Subjects
Male ,0301 basic medicine ,Time Factors ,Histology ,Confocal ,Fluorescent Antibody Technique ,Immunofluorescence ,Article ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Imaging, Three-Dimensional ,Lectins ,medicine ,Animals ,Sodium dodecyl sulfate ,Acrylamide ,Microscopy, Confocal ,Chromatography ,Dose-Response Relationship, Drug ,Staining and Labeling ,medicine.diagnostic_test ,biology ,General Neuroscience ,Brain ,Lectin ,Fluorescence ,Staining ,Mice, Inbred C57BL ,Parvalbumins ,030104 developmental biology ,Monomer ,chemistry ,biology.protein ,Anatomy - Abstract
The CLARITY technique enables three-dimensional visualization of fluorescent-labeled biomolecules in clarified intact brain samples, affording a unique view of molecular neuroanatomy and neurocircuitry. It is therefore, essential to find the ideal combination for clearing tissue and detecting the fluorescent-labeled signal. This method requires the formation of a formaldehyde-acrylamide fixative-generated hydrogel mesh through which cellular lipid is removed with sodium dodecyl sulfate. Several laboratories have used differential acrylamide and detergent concentrations to achieve better tissue clearing and antibody penetration, but the potential effects upon fluorescent signal retention is largely unknown. In an effort to optimize CLARITY processing procedures we performed quantitative parvalbumin immunofluorescence and lectin-based vasculature staining using either 4 or 8% sodium dodecyl sulfate detergent in combination with different acrylamide formulas in mouse brain slices. Using both confocal and CLARITY-optimized lightsheet microscope-acquired images, we demonstrate that 2% acrylamide monomer combined with 0.0125% bis-acrylamide and cleared with 4% sodium dodecyl sulfate generally provides the most optimal signal visualization amongst various hydrogel monomer concentrations, lipid removal times, and detergent concentrations.
- Published
- 2017
- Full Text
- View/download PDF
39. Effect of Early-Life Fluoxetine on Anxiety-Like Behaviors in BDNF Val66Met Mice
- Author
-
Deqiang Jing, Helena Freilingsdorf, Chienchun Huang, Francis S. Lee, Barbara L. Hempstead, B. J. Casey, Charles E. Glatt, Jianmin Yang, Tina Marinic, Anfei Li, Iva Dincheva, and Kevin G. Bath
- Subjects
Dorsal Raphe Nucleus ,Male ,0301 basic medicine ,medicine.medical_specialty ,medicine.medical_treatment ,Prefrontal Cortex ,S100 Calcium Binding Protein beta Subunit ,Anxiety ,Polymorphism, Single Nucleotide ,Article ,Eating ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Neurochemical ,Neurotrophic factors ,Fluoxetine ,Internal medicine ,medicine ,Animals ,Gene Knock-In Techniques ,Maze Learning ,Brain-Derived Neurotrophic Factor ,Growth factor ,Age Factors ,Fear ,Phenotype ,Psychiatry and Mental health ,030104 developmental biology ,Endocrinology ,Anti-Anxiety Agents ,Mechanism of action ,medicine.symptom ,Psychology ,rs6265 ,Selective Serotonin Reuptake Inhibitors ,030217 neurology & neurosurgery ,Serotonergic Neurons ,Clinical psychology ,medicine.drug - Abstract
Adolescence is a developmental stage in which the incidence of psychiatric disorders, such as anxiety disorders, peaks. Selective serotonin reuptake inhibitors (SSRIs) are the main class of agents used to treat anxiety disorders. However, the impact of SSRIs on the developing brain during adolescence remains unknown. The authors assessed the impact of developmentally timed SSRI administration in a genetic mouse model displaying elevated anxiety-like behaviors.Knock-in mice containing a common human single-nucleotide polymorphism (Val66Met; rs6265) in brain-derived neurotrophic factor (BDNF), a growth factor implicated in the mechanism of action of SSRIs, were studied based on their established phenotype of increased anxiety-like behavior. Timed administration of fluoxetine was delivered during one of three developmental periods (postnatal days 21-42, 40-61, or 60-81), spanning the transition from childhood to adulthood. Neurochemical and anxiety-like behavioral analyses were performed.We identified a "sensitive period" during periadolescence (postnatal days 21-42) in which developmentally timed fluoxetine administration rescued anxiety-like phenotypes in BDNF Val66Met mice in adulthood. Compared with littermate controls, BDNFThese findings suggest that SSRI administration during a "sensitive period" during periadolescence leads to long-lasting anxiolytic effects in a genetic mouse model of elevated anxiety-like behaviors. These persistent effects highlight the role of BDNF in the maturation of the serotonin system and the capacity to enhance its development through a pharmacological intervention.
- Published
- 2017
- Full Text
- View/download PDF
40. BDNF at the synapse: why location matters
- Author
-
Keri Martinowich, Minseok Song, and Francis S. Lee
- Subjects
0301 basic medicine ,Context (language use) ,Biology ,Article ,Synapse ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Mediator ,Neurotrophic factors ,Neuroplasticity ,Animals ,Humans ,Epigenetics ,Molecular Biology ,Neurons ,Brain-derived neurotrophic factor ,Neuronal Plasticity ,Brain-Derived Neurotrophic Factor ,Brain ,Antidepressive Agents ,Psychiatry and Mental health ,030104 developmental biology ,nervous system ,Synapses ,Ketamine ,Signal transduction ,Neuroscience ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Neurotrophic factors, a family of secreted proteins that support the growth, survival and differentiation of neurons, have been intensively studied for decades due to the powerful and diverse effects on neuronal physiology, as well as their therapeutic potential. Such efforts have led to a detailed understanding on the molecular mechanisms of neurotrophic factor signaling. One member, brain-derived neurotrophic factor (BDNF) has drawn much attention due to its pleiotropic roles in the central nervous system and implications in various brain disorders. In addition, recent advances linking the rapid-acting antidepressant, ketamine, to BDNF translation and BDNF-dependent signaling, has re-emphasized the importance of understanding the precise details of BDNF biology at the synapse. Although substantial knowledge related to the genetic, epigenetic, cell biological and biochemical aspects of BDNF biology has now been established, certain aspects related to the precise localization and release of BDNF at the synapse have remained obscure. A recent series of genetic and cell biological studies have shed light on the question-the site of BDNF release at the synapse. In this Perspectives article, these new insights will be placed in the context of previously unresolved issues related to BDNF biology, as well as how BDNF may function as a downstream mediator of newer pharmacological agents currently under investigation for treating psychiatric disorders.
- Published
- 2017
- Full Text
- View/download PDF
41. The Role of the Endocannabinoid System and Genetic Variation in Adolescent Brain Development
- Author
-
Heidi C. Meyer, Dylan G. Gee, and Francis S. Lee
- Subjects
0301 basic medicine ,Brain development ,Adolescent ,Emotions ,Neurotransmission ,03 medical and health sciences ,0302 clinical medicine ,Genetic variation ,Neuropsychopharmacology Reviews ,medicine ,Biological neural network ,Animals ,Humans ,Sexual Maturation ,Prefrontal cortex ,Pharmacology ,Brain ,Genetic Variation ,Cognition ,Adolescent Development ,Endocannabinoid system ,Psychiatry and Mental health ,030104 developmental biology ,Anxiety ,medicine.symptom ,Psychology ,Neuroscience ,030217 neurology & neurosurgery ,Endocannabinoids - Abstract
During adolescence, both rodent and human studies have revealed dynamic changes in the developmental trajectories of corticolimbic structures, which are known to contribute to the regulation of fear and anxiety-related behaviors. The endocannabinoid (eCB) system critically regulates stress responsivity and anxiety throughout the life span. Emerging evidence suggests that during adolescence, changes in eCB signaling contribute to the maturation of local and corticolimbic circuit populations of neurons, such as mediating the balance between excitatory and inhibitory neurotransmission within the prefrontal cortex. This function of the eCB system facilitates efficient communication within and between brain regions and serves a central role in establishing complex and adaptive cognitive and behavioral processing. Although these peri-adolescent changes in eCB signaling promote brain development and plasticity, they also render this period a particularly sensitive one for environmental perturbations to these normative fluctuations in eCB signaling, such as stress, potentially leading to altered developmental trajectories of neural circuits governing emotional behaviors. In this review, we focus on the role of eCB signaling on the regulation of stress and anxiety-related behaviors both during and after adolescence. Moreover, we discuss the functional implications of human genetic variation in the eCB system for the risk for anxiety and consequences of stress across development and into adulthood.
- Published
- 2017
- Full Text
- View/download PDF
42. D-cycloserine augmentation of exposure-based cognitive behavior therapy for anxiety, obsessive-compulsive, and posttraumatic stress disorders: A systematic review and meta-analysis of individual participant data
- Author
-
David F. Tolin, Page L. Anderson, Anja Siewert-Siegmund, Paolo Frumento, Daniel A. Geller, Michael W. Otto, Agnes van Minnen, Eric J. Lenze, Tanya K. Murphy, Benedetta Monzani, Cassidy A. Gutner, David Mataix-Cols, Lorena Fernández de la Cruz, Thomas L. Rodebaugh, Ana Pérez-Vigil, Stefan G. Hofmann, Boadie W. Dunlop, Isobel Heyman, Seth D. Norrholm, Mark H. Pollack, Maryrose Gerardi, Joseph P. H. McNamara, Claudia Finck, Katarzyna Wyka, Tanja Jovanovic, Carl F. Weems, Judith Cukor, Cheri A. Levinson, Matt G. Kushner, Gary R. Geffken, David Rosenfield, Adam J. Guastella, Jens Plag, Gert-Jan Hendriks, Allison M. Waters, Sabine Wilhelm, Fabian Golfels, Jasper A. J. Smits, Lara J. Farrell, Barbara O. Rothbaum, Adam B. Lewin, JoAnn Difede, Wayne K. Goodman, Harry McConnell, Rianne A. de Kleine, Andreas Ströhle, Christian Rück, Francis S. Lee, Erik Andersson, Paul Thuras, Michael S. Scheeringa, Kerry J. Ressler, Eric A. Storch, Margaret Altemus, Michael Davis, and Candyce D. Tart
- Subjects
Obsessive-Compulsive Disorder ,N-Methylaspartate ,medicine.medical_treatment ,Stress-related disorders Donders Center for Medical Neuroscience [Radboudumc 13] ,Placebo-controlled study ,Implosive Therapy ,Specific phobia ,Stress Disorders, Post-Traumatic ,Experimental Psychopathology and Treatment ,03 medical and health sciences ,0302 clinical medicine ,Outcome Assessment, Health Care ,Excitatory Amino Acid Agonists ,medicine ,Humans ,10. No inequality ,Panic disorder ,Social anxiety ,Repeated measures design ,Drug Synergism ,medicine.disease ,Anxiety Disorders ,Combined Modality Therapy ,Antidepressive Agents ,030227 psychiatry ,3. Good health ,Cognitive behavioral therapy ,Psychiatry and Mental health ,Cycloserine ,Anxiety ,medicine.symptom ,Psychology ,030217 neurology & neurosurgery ,Clinical psychology ,Agoraphobia - Abstract
Contains fulltext : 174490.pdf (Publisher’s version ) (Open Access) Importance: Whether and under which conditions D-cycloserine (DCS) augments the effects of exposure-based cognitive behavior therapy for anxiety, obsessive-compulsive, and posttraumatic stress disorders is unclear. Objective: To clarify whether DCS is superior to placebo in augmenting the effects of cognitive behavior therapy for anxiety, obsessive-compulsive, and posttraumatic stress disorders and to evaluate whether antidepressants interact with DCS and the effect of potential moderating variables. Data Sources: PubMed, EMBASE, and PsycINFO were searched from inception to February 10, 2016. Reference lists of previous reviews and meta-analyses and reports of randomized clinical trials were also checked. Study Selection: Studies were eligible for inclusion if they were (1) double-blind randomized clinical trials of DCS as an augmentation strategy for exposure-based cognitive behavior therapy and (2) conducted in humans diagnosed as having specific phobia, social anxiety disorder, panic disorder with or without agoraphobia, obsessive-compulsive disorder, or posttraumatic stress disorder. Data Extraction and Synthesis: Raw data were obtained from the authors and quality controlled. Data were ranked to ensure a consistent metric across studies (score range, 0-100). We used a 3-level multilevel model nesting repeated measures of outcomes within participants, who were nested within studies. Results: Individual participant data were obtained for 21 of 22 eligible trials, representing 1047 of 1073 eligible participants. When controlling for antidepressant use, participants receiving DCS showed greater improvement from pretreatment to posttreatment (mean difference, -3.62; 95% CI, -0.81 to -6.43; P = .01; d = -0.25) but not from pretreatment to midtreatment (mean difference, -1.66; 95% CI, -4.92 to 1.60; P = .32; d = -0.14) or from pretreatment to follow-up (mean difference, -2.98, 95% CI, -5.99 to 0.03; P = .05; d = -0.19). Additional analyses showed that participants assigned to DCS were associated with lower symptom severity than those assigned to placebo at posttreatment and at follow-up. Antidepressants did not moderate the effects of DCS. None of the prespecified patient-level or study-level moderators was associated with outcomes. Conclusions and Relevance: D-cycloserine is associated with a small augmentation effect on exposure-based therapy. This effect is not moderated by the concurrent use of antidepressants. Further research is needed to identify patient and/or therapy characteristics associated with DCS response. 10 p.
- Published
- 2017
43. Ventral hippocampus interacts with prelimbic cortex during inhibition of threat response via learned safety in both mice and humans
- Author
-
Sadie J. Zacharek, Francis S. Lee, Baila S. Hall, Anfei Li, Heidi C. Meyer, Jeffrey D. Mandell, Dylan G. Gee, Conor Liston, Ruirong Yang, Jason T. Haberman, Paola Odriozola, and Emily M. Cohodes
- Subjects
Multidisciplinary ,business.industry ,Infralimbic cortex ,Ventromedial prefrontal cortex ,Hippocampus ,Extinction (psychology) ,Amygdala ,Freezing behavior ,medicine.anatomical_structure ,PNAS Plus ,medicine ,business ,Neuroscience ,Anterior cingulate cortex ,Basolateral amygdala - Abstract
Heightened fear and inefficient safety learning are key features of fear and anxiety disorders. Evidence-based interventions for anxiety disorders, such as cognitive behavioral therapy, primarily rely on mechanisms of fear extinction. However, up to 50% of clinically anxious individuals do not respond to current evidence-based treatment, suggesting a critical need for new interventions based on alternative neurobiological pathways. Using parallel human and rodent conditioned inhibition paradigms alongside brain imaging methodologies, we investigated neural activity patterns in the ventral hippocampus in response to stimuli predictive of threat or safety and compound cues to test inhibition via safety in the presence of threat. Distinct hippocampal responses to threat, safety, and compound cues suggest that the ventral hippocampus is involved in conditioned inhibition in both mice and humans. Moreover, unique response patterns within target-differentiated subpopulations of ventral hippocampal neurons identify a circuit by which fear may be inhibited via safety. Specifically, ventral hippocampal neurons projecting to the prelimbic cortex, but not to the infralimbic cortex or basolateral amygdala, were more active to safety and compound cues than threat cues, and activity correlated with freezing behavior in rodents. A corresponding distinction was observed in humans: hippocampal–dorsal anterior cingulate cortex functional connectivity—but not hippocampal–anterior ventromedial prefrontal cortex or hippocampal–basolateral amygdala connectivity—differentiated between threat, safety, and compound conditions. These findings highlight the potential to enhance treatment for anxiety disorders by targeting an alternative neural mechanism through safety signal learning.
- Published
- 2019
44. D3 dopamine receptors and a missense mutation of fatty acid amide hydrolase linked in mouse and men: implication for addiction
- Author
-
Matthew N. Hill, Esmaeil Mansouri, Patricia Di Ciano, Bernard Le Foll, Isabelle Boileau, Georgia Balsevich, Rachel F. Tyndale, Stephen J. Kish, Junchao Tong, José N. Nobrega, Mathew E. Sloan, Christian S. Hendershot, Francis S. Lee, and Laura M. Best
- Subjects
Adult ,Male ,medicine.medical_specialty ,Substance-Related Disorders ,Mutation, Missense ,In situ hybridization ,Polymorphism, Single Nucleotide ,Article ,Amidohydrolases ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Young Adult ,0302 clinical medicine ,Fatty acid amide hydrolase ,Dopamine receptor D3 ,Internal medicine ,medicine ,Animals ,Humans ,Gene Knock-In Techniques ,RNA, Messenger ,Aged ,Pharmacology ,Chemistry ,Dopaminergic ,Receptors, Dopamine D3 ,Brain ,Anandamide ,Middle Aged ,Endocannabinoid system ,030227 psychiatry ,Psychiatry and Mental health ,Endocrinology ,nervous system ,Dopamine receptor ,Positron-Emission Tomography ,Islands of Calleja ,Autoradiography ,lipids (amino acids, peptides, and proteins) ,Female ,030217 neurology & neurosurgery - Abstract
The endocannabinoid and dopaminergic systems have independently been implicated in substance use disorder and obesity. We investigated a potential interaction between genetically inherited variation in fatty acid amide hydrolase (FAAH, C385A), which metabolizes the cannabis-like endocannabinoid anandamide, and dopaminergic system, measured by dopamine receptor levels and mRNA. Binding of the dopamine D3 preferring probe [C-11]-(+)-PHNO was measured with positron emission tomography (PET) in 79 human subjects genotyped for the FAAH C385A polymorphism (36/79 AC + AA). Autoradiography with [H-3]-(+)-PHNO and in situ hybridization with a D3-specific S-35 riboprobe were carried out in 30 knock-in mice with the FAAH C385A polymorphism (20/30 AC + AA). We found that the FAAH genetic variant C385A was associated with significantly higher (+)-PHNO binding in both humans and in knock-in mice, and this effect was restricted to D3 selective brain regions (limbic striatum, globus pallidus, and ventral pallidum (9–14%; p
- Published
- 2019
45. Physical Exercise Affects Adipose Tissue Profile and Prevents Arterial Thrombosis in
- Author
-
Leonardo, Sandrini, Alessandro, Ieraci, Patrizia, Amadio, Marta, Zarà, Nico, Mitro, Francis S, Lee, Elena, Tremoli, and Silvia Stella, Barbieri
- Subjects
Inflammation ,Adipose Tissue, White ,Brain-Derived Neurotrophic Factor ,Thrombosis ,Arteries ,arterial thrombosis ,Polymorphism, Single Nucleotide ,Article ,Cell Line ,adipose tissue ,adipogenesis ,Mice ,BDNF ,physical exercise ,Physical Conditioning, Animal ,Adipocytes ,Val66Met polymorphism ,Animals ,Gene Knock-In Techniques - Abstract
Adipose tissue accumulation is an independent and modifiable risk factor for cardiovascular disease (CVD). The recent CVD European Guidelines strongly recommend regular physical exercise (PE) as a management strategy for prevention and treatment of CVD associated with metabolic disorders and obesity. Although mutations as well as common genetic variants, including the brain-derived neurotrophic factor (BDNF) Val66Met polymorphism, are associated with increased body weight, eating and neuropsychiatric disorders, and myocardial infarction, the effect of this polymorphism on adipose tissue accumulation and regulation as well as its relation to obesity/thrombosis remains to be elucidated. Here, we showed that white adipose tissue (WAT) of humanized knock-in BDNFVal66Met (BDNFMet/Met) mice is characterized by an altered morphology and an enhanced inflammatory profile compared to wild-type BDNFVal/Val. Four weeks of voluntary PE restored the adipocyte size distribution, counteracted the inflammatory profile of adipose tissue, and prevented the prothrombotic phenotype displayed, per se, by BDNFMet/Met mice. C3H10T1/2 cells treated with the Pro-BDNFMet peptide well recapitulated the gene alterations observed in BDNFMet/Met WAT mice. In conclusion, these data indicate the strong impact of lifestyle, in particular of the beneficial effect of PE, on the management of arterial thrombosis and inflammation associated with obesity in relation to the specific BDNF Val66Met mutation.
- Published
- 2019
46. Cannabis and the Developing Brain: Insights into Its Long-Lasting Effects
- Author
-
Miriam Melis, Francis S. Lee, Yasmin L. Hurd, Sagnik Bhattacharyya, Mikhail V. Pletnikov, and Olivier J. Manzoni
- Subjects
0301 basic medicine ,Long lasting ,medicine.medical_specialty ,Adolescent ,03 medical and health sciences ,0302 clinical medicine ,Cognition ,Pregnancy ,Epidemiology ,medicine ,Neural system ,Animals ,Humans ,Psychiatry ,Cannabis ,biology ,Human studies ,business.industry ,General Neuroscience ,Mental Disorders ,Symposium and Mini-Symposium ,biology.organism_classification ,Mental health ,030104 developmental biology ,Prenatal Exposure Delayed Effects ,Female ,Marijuana Use ,Substance use ,Erratum ,business ,030217 neurology & neurosurgery - Abstract
The recent shift in sociopolitical debates and growing liberalization of cannabis use across the globe has raised concern regarding its impact on vulnerable populations, such as pregnant women and adolescents. Epidemiological studies have long demonstrated a relationship between developmental cannabis exposure and later mental health symptoms. This relationship is especially strong in people with particular genetic polymorphisms, suggesting that cannabis use interacts with genotype to increase mental health risk. Seminal animal research directly linked prenatal and adolescent exposure to delta-9-tetrahydrocannabinol, the major psychoactive component of cannabis, with protracted effects on adult neural systems relevant to psychiatric and substance use disorders. In this article, we discuss some recent advances in understanding the long-term molecular, epigenetic, electrophysiological, and behavioral consequences of prenatal, perinatal, and adolescent exposure to cannabis/delta-9-tetrahydrocannabinol. Insights are provided from both animal and human studies, includingin vivoneuroimaging strategies.
- Published
- 2019
47. Enhanced exposure therapy for combat-related Posttraumatic Stress Disorder (PTSD): Study protocol for a randomized controlled trial
- Author
-
Seth D. Norrholm, Albert Rizzo, Lisa Spielman, Tanja Jovanovic, Michael J. Roy, Christopher Reist, Charles E. Glatt, Katarzyna Wyka, Barbara O. Rothbaum, Francis S. Lee, and JoAnn Difede
- Subjects
medicine.medical_specialty ,Prolonged exposure therapy ,business.industry ,medicine.medical_treatment ,Virtual Reality Exposure Therapy ,Exposure therapy ,Implosive Therapy ,Cognition ,General Medicine ,Extinction (psychology) ,Placebo ,Combined Modality Therapy ,law.invention ,Stress Disorders, Post-Traumatic ,Randomized controlled trial ,law ,Cycloserine ,Intervention (counseling) ,Emergency medicine ,medicine ,Humans ,Pharmacology (medical) ,business ,Veterans - Abstract
Background PTSD, which has been identified in up to 23% of post-9-11 veterans, often results in a chronic, pernicious course. Thus, effective treatments are imperative. The Institute of Medicine (IOM) concluded that the only intervention for PTSD with sufficient evidence to conclude efficacy is exposure therapy. This Phase III trial compares the efficacy of exposure therapy for combat-related PTSD delivered in two different formats- via virtual reality exposure therapy (VRE) or prolonged exposure therapy (PE)- combined with D-Cycloserine (DCS), a cognitive enhancer shown to facilitate the extinction of fear. Methods/design Military personnel of any duty status and civilians deployed to Iraq and Afghanistan were eligible. Participants were randomly assigned to 9 sessions of exposure therapy (VRE or PE) and medication (50 mg DCS or placebo). Participants were treated at three geographically diverse sites. Participants were re-assessed at 3-months post-treatment. The co-primary hypotheses are that (1) DCS will augment response to exposure therapy (both VRE and PE) on PTSD symptoms; (2) VRE will be associated with greater improvement than PE. Genetic and psychophysiological markers will be evaluated as potential moderators and mediators of treatment outcomes as well as secondary outcomes. Discussion This study is the first to compare the relative efficacy of DCS-augmented VRE versus PE on PTSD symptoms. The design has several advantages: participants received an active, effective treatment and predictors of response to treatment included genetic and psychobiological measures. The results may directly influence the future delivery of services, and contribute to the development of a standardized treatment protocol. Trial registration: NCT01352637 .
- Published
- 2019
48. Diminished Fear Extinction in Adolescents Is Associated With an Altered Somatostatin Interneuron-Mediated Inhibition in the Infralimbic Cortex
- Author
-
Francis S. Lee, Christopher Galvin, Peter Koppensteiner, Richard Von Itter, Ipe Ninan, and Riccardo Melani
- Subjects
0301 basic medicine ,Male ,Adolescent ,Interneuron ,Infralimbic cortex ,Prefrontal Cortex ,Anxiety ,Biology ,Article ,Extinction, Psychological ,03 medical and health sciences ,Glutamatergic ,Mice ,0302 clinical medicine ,Interneurons ,medicine ,Limbic System ,Humans ,Animals ,Fear conditioning ,Prefrontal cortex ,Biological Psychiatry ,Neuronal Plasticity ,Pyramidal Cells ,Glutamate receptor ,Extinction (psychology) ,Fear ,Optogenetics ,Inhibition, Psychological ,030104 developmental biology ,medicine.anatomical_structure ,Synapses ,Neuron ,Somatostatin ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Background Rodents and humans show an attenuation of fear extinction during adolescence, which coincides with the onset of several psychiatric disorders. Although the ethological relevance and the underlying mechanism are largely unknown, the suppression of fear extinction during adolescence is associated with a diminished plasticity in the glutamatergic neurons of the infralimbic medial prefrontal cortex, a brain region critical for fear extinction. Given the putative effect of synaptic inhibition on glutamatergic neuron activity, we studied whether gamma-aminobutyric acidergic neurons in the infralimbic medial prefrontal cortex are involved in the suppression of fear extinction during adolescence. Methods We assessed membrane and synaptic properties in parvalbumin-positive interneurons (PVINs) and somatostatin-positive interneurons (SSTINs) in male preadolescent, adolescent, and adult mice. The effect of fear conditioning and extinction on PVIN-pyramidal neuron and SSTIN-pyramidal neuron synapses in male preadolescent, adolescent, and adult mice was evaluated using an optogenetic approach. Results The development of the membrane excitability of PVINs is delayed and reaches maturity only by adulthood, while the SSTIN membrane properties are developed early and remain stable during development from preadolescence to adulthood. Although the synaptic inhibition mediated by PVINs undergoes a protracted development, it does not exhibit a fear behavior–specific plasticity. However, the synaptic inhibition mediated by SSTINs undergoes an adolescence-specific enhancement, and this increased inhibition is suppressed by fear learning but is not restored by extinction training. This altered plasticity during adolescence overlapped with a reduction in calcium-permeable glutamate receptors in SSTINs. Conclusions The adolescence-specific plasticity in the SSTINs might play a role in fear extinction suppression during adolescence in mice.
- Published
- 2019
49. Using a Developmental Ecology Framework to Align Fear Neurobiology Across Species
- Author
-
Maya Opendak, Anfei Li, Nim Tottenham, Chelsea Harmon, Bridget L. Callaghan, Heidi C. Meyer, Michelle Van Tieghem, Francis S. Lee, and Regina M. Sullivan
- Subjects
Ecology (disciplines) ,Context (language use) ,050105 experimental psychology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Child Development ,Animals ,Humans ,Learning ,0501 psychology and cognitive sciences ,Fear learning ,Parent-Child Relations ,Child ,Human studies ,Behavior, Animal ,Ecology ,05 social sciences ,General Medicine ,Fear ,Object Attachment ,Psychiatry and Mental health ,Clinical Psychology ,Increased risk ,Psychology ,Neuroscience ,030217 neurology & neurosurgery ,Psychopathology - Abstract
Children's development is largely dependent on caregiving; when caregiving is disrupted, children are at increased risk for numerous poor outcomes, in particular psychopathology. Therefore, determining how caregivers regulate children's affective neurobiology is essential for understanding psychopathology etiology and prevention. Much of the research on affective functioning uses fear learning to map maturation trajectories, with both rodent and human studies contributing knowledge. Nonetheless, as no standard framework exists through which to interpret developmental effects across species, research often remains siloed, thus contributing to the current therapeutic impasse. Here, we propose a developmental ecology framework that attempts to understand fear in the ecological context of the child: their relationship with their parent. By referring to developmental goals that are shared across species (to attach to, then, ultimately, separate from the parent), this framework provides a common grounding from which fear systems and their dysfunction can be understood, thus advancing research on psychopathologies and their treatment.
- Published
- 2019
50. Endocannabinoid Signaling Collapse Mediates Stress-Induced Amygdalo-Cortical Strengthening
- Author
-
Sachin Patel, David J. Marcus, Gaurav Bedse, Jennifer Urbano Blackford, Andrew D. Gaulden, Eric Delpire, Francis S. Lee, Jacqueline A. Clauss, and James D. Ryan
- Subjects
Mechanism (biology) ,2-Arachidonoylglycerol ,Glutamate receptor ,Translation (biology) ,Biology ,Endocannabinoid system ,Amygdala ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,medicine ,Prefrontal cortex ,Neuroscience ,Basolateral amygdala - Abstract
Functional coupling between the basolateral amygdala (BLA) and areas of the dorsal prefrontal cortex (dPFC) have been implicated in the generation of negative affective states under stressful conditions; however, the synaptic and molecular mechanisms by which stress increases BLA-dPFC synaptic strength and generates anxiety-like behaviors are not well understood. Here we show that BLA-dPFC functional connectivity is enhanced under conditions of potential threat and correlates with trait anxiety in humans and that acute stress exposure increases anxiety-like behavior and synaptic strength within a reciprocal BLA-dPFC-BLA subcircuit in mice. Importantly, we identify 2-arachidonoylglycerol (2-AG)-mediated endocannabinoid signaling as a key mechanism limiting glutamate release at BLA-dPFC synapses and the collapse of multimodal 2-AG signaling as a molecular mechanism leading to persistent circuit-specific synaptic strengthening and anxiety-like behaviors after stress exposure. These data suggest circuit-specific impairment in 2-AG signaling could facilitate functional coupling between the BLA and dPFC and the translation of environmental stress to affective pathology.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.