Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 174.196
Filtrar
Mais filtros

Temas
Intervalo de ano de publicação
1.
Cell ; 186(3): 560-576.e17, 2023 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-36693374

RESUMO

Downward social mobility is a well-known mental risk factor for depression, but its neural mechanism remains elusive. Here, by forcing mice to lose against their subordinates in a non-violent social contest, we lower their social ranks stably and induce depressive-like behaviors. These rank-decline-associated depressive-like behaviors can be reversed by regaining social status. In vivo fiber photometry and single-unit electrophysiological recording show that forced loss, but not natural loss, generates negative reward prediction error (RPE). Through the lateral hypothalamus, the RPE strongly activates the brain's anti-reward center, the lateral habenula (LHb). LHb activation inhibits the medial prefrontal cortex (mPFC) that controls social competitiveness and reinforces retreats in contests. These results reveal the core neural mechanisms mutually promoting social status loss and depressive behaviors. The intertwined neuronal signaling controlling mPFC and LHb activities provides a mechanistic foundation for the crosstalk between social mobility and psychological disorder, unveiling a promising target for intervention.


Assuntos
Habenula , Status Social , Camundongos , Animais , Recompensa , Comportamento Social , Habenula/fisiologia , Depressão
2.
Cell ; 185(1): 1-3, 2022 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-34995512

RESUMO

Psychiatric disease is one of the greatest health challenges of our time. The pipeline for conceptually novel therapeutics remains low, in part because uncovering the biological mechanisms of psychiatric disease has been difficult. We asked experts researching different aspects of psychiatric disease: what do you see as the major urgent questions that need to be addressed? Where are the next frontiers, and what are the current hurdles to understanding the biological basis of psychiatric disease?


Assuntos
Antidepressivos/uso terapêutico , Ciência de Dados/métodos , Depressão/tratamento farmacológico , Depressão/metabolismo , Transtorno Depressivo/tratamento farmacológico , Transtorno Depressivo/metabolismo , Genômica/métodos , Medicina de Precisão/métodos , Pesquisa Translacional Biomédica/métodos , Animais , Depressão/genética , Transtorno Depressivo/genética , Humanos , Neurônios/metabolismo , Córtex Pré-Frontal/metabolismo , Resultado do Tratamento
3.
Immunity ; 57(4): 837-839, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38599175

RESUMO

Activation of the peripheral immune system contributes to stress-related neuropsychiatric symptoms. Recently in Nature, Cathomas et al. demonstrate that stress-induced social avoidance is mediated by monocyte-derived MMP8 that remodels the extracellular space of the nucleus accumbens.


Assuntos
Depressão , Monócitos , Estresse Psicológico , Núcleo Accumbens
4.
Cell ; 173(1): 166-180.e14, 2018 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-29502969

RESUMO

Brain-wide fluctuations in local field potential oscillations reflect emergent network-level signals that mediate behavior. Cracking the code whereby these oscillations coordinate in time and space (spatiotemporal dynamics) to represent complex behaviors would provide fundamental insights into how the brain signals emotional pathology. Using machine learning, we discover a spatiotemporal dynamic network that predicts the emergence of major depressive disorder (MDD)-related behavioral dysfunction in mice subjected to chronic social defeat stress. Activity patterns in this network originate in prefrontal cortex and ventral striatum, relay through amygdala and ventral tegmental area, and converge in ventral hippocampus. This network is increased by acute threat, and it is also enhanced in three independent models of MDD vulnerability. Finally, we demonstrate that this vulnerability network is biologically distinct from the networks that encode dysfunction after stress. Thus, these findings reveal a convergent mechanism through which MDD vulnerability is mediated in the brain.


Assuntos
Encéfalo/fisiologia , Depressão/patologia , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Depressão/fisiopatologia , Modelos Animais de Doenças , Estimulação Elétrica , Eletrodos Implantados , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Ketamina/farmacologia , Aprendizado de Máquina , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fenômenos Fisiológicos/efeitos dos fármacos , Córtex Pré-Frontal/fisiologia , Estresse Psicológico
5.
Immunity ; 56(3): 469-471, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36921572

RESUMO

Investigators have long suspected a link between inflammation and depression, but the underlying mechanisms are not well understood. Fang et al. report that lipopolysaccharide-binding protein regulates monoamine biosynthesis and might be a missing link and potential therapeutic target for inflammation-associated depressive behaviors.


Assuntos
Proteínas de Transporte , Depressão , Humanos , Proteínas de Fase Aguda , Inflamação
6.
Immunity ; 56(3): 620-634.e11, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36854305

RESUMO

Monoamine insufficiency is suggested to be associated with depressive features such as sadness, anhedonia, insomnia, and cognitive dysfunction, but the mechanisms that cause it are unclear. We found that the acute-phase protein lipopolysaccharide-binding protein (LBP) inhibits monoamine biosynthesis by acting as an endogenous inhibitor of dopamine-ß-hydroxylase (DBH) and aromatic-L-amino-acid-decarboxylase (DDC). LBP expression was increased in individuals with depression and by diverse stress challenges in mice. LBP antibodies and LBP knockdown inhibited monoamine insufficiency and depression-like features in mice, which worsened with LBP overexpression or administration. Monoamine insufficiency and depression-like symptoms were not induced by stressful stimuli in LBP-deficient mice, further highlighting a role for LBP in stress-induced depression, and a peptide we designed that blocks LBP-DBH and LBP-DDC interactions showed anti-depression effects in mice. This study reveals an important role for LBP in regulating monoamine biosynthesis and suggests that targeting LBP may have potential as a treatment for some individuals with depression.


Assuntos
Proteínas de Transporte , Depressão , Camundongos , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/metabolismo , Glicoproteínas de Membrana/metabolismo , Aminas
7.
Cell ; 170(2): 284-297.e18, 2017 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-28689640

RESUMO

Major depressive disorder (MDD) patients display a common but often variable set of symptoms making successful, sustained treatment difficult to achieve. Separate depressive symptoms may be encoded by differential changes in distinct circuits in the brain, yet how discrete circuits underlie behavioral subsets of depression and how they adapt in response to stress has not been addressed. We identify two discrete circuits of parvalbumin-positive (PV) neurons in the ventral pallidum (VP) projecting to either the lateral habenula or ventral tegmental area contributing to depression. We find that these populations undergo different electrophysiological adaptations in response to social defeat stress, which are normalized by antidepressant treatment. Furthermore, manipulation of each population mediates either social withdrawal or behavioral despair, but not both. We propose that distinct components of the VP PV circuit can subserve related, yet separate depressive-like phenotypes in mice, which could ultimately provide a platform for symptom-specific treatments of depression.


Assuntos
Prosencéfalo Basal/fisiopatologia , Depressão/patologia , Neurônios/patologia , Animais , Aprendizagem da Esquiva , Prosencéfalo Basal/patologia , Depressão/fisiopatologia , Transtorno Depressivo Maior/patologia , Transtorno Depressivo Maior/fisiopatologia , Feminino , Técnicas In Vitro , Masculino , Mesencéfalo/metabolismo , Mesencéfalo/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Parvalbuminas/metabolismo
8.
Annu Rev Neurosci ; 46: 341-358, 2023 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-37018916

RESUMO

The field of stereotactic neurosurgery developed more than 70 years ago to address a therapy gap for patients with severe psychiatric disorders. In the decades since, it has matured tremendously, benefiting from advances in clinical and basic sciences. Deep brain stimulation (DBS) for severe, treatment-resistant psychiatric disorders is currently poised to transition from a stage of empiricism to one increasingly rooted in scientific discovery. Current drivers of this transition are advances in neuroimaging, but rapidly emerging ones are neurophysiological-as we understand more about the neural basis of these disorders, we will more successfully be able to use interventions such as invasive stimulation to restore dysfunctional circuits to health. Paralleling this transition is a steady increase in the consistency and quality of outcome data. Here, we focus on obsessive-compulsive disorder and depression, two topics that have received the most attention in terms of trial volume and scientific effort.


Assuntos
Estimulação Encefálica Profunda , Transtorno Obsessivo-Compulsivo , Humanos , Estimulação Encefálica Profunda/métodos , Depressão , Procedimentos Neurocirúrgicos/métodos , Transtorno Obsessivo-Compulsivo/cirurgia , Neuroimagem
9.
Annu Rev Neurosci ; 45: 581-601, 2022 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-35508195

RESUMO

Depression is an episodic form of mental illness characterized by mood state transitions with poorly understood neurobiological mechanisms. Antidepressants reverse the effects of stress and depression on synapse function, enhancing neurotransmission, increasing plasticity, and generating new synapses in stress-sensitive brain regions. These properties are shared to varying degrees by all known antidepressants, suggesting that synaptic remodeling could play a key role in depression pathophysiology and antidepressant function. Still, it is unclear whether and precisely how synaptogenesis contributes to mood state transitions. Here, we review evidence supporting an emerging model in which depression is defined by a distinct brain state distributed across multiple stress-sensitive circuits, with neurons assuming altered functional properties, synapse configurations, and, importantly, a reduced capacity for plasticity and adaptation. Antidepressants act initially by facilitating plasticity and enabling a functional reconfiguration of this brain state. Subsequently, synaptogenesis plays a specific role in sustaining these changes over time.


Assuntos
Antidepressivos , Depressão , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Plasticidade Neuronal/fisiologia , Neurônios , Sinapses/fisiologia , Transmissão Sináptica/fisiologia
10.
Immunity ; 54(2): 225-234.e6, 2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33476547

RESUMO

Microglia are activated in many neurological diseases and have been suggested to play an important role in the development of affective disorders including major depression. To investigate how microglial signaling regulates mood, we used bidirectional chemogenetic manipulations of microglial activity in mice. Activation of microglia in the dorsal striatum induced local cytokine expression and a negative affective state characterized by anhedonia and aversion, whereas inactivation of microglia blocked aversion induced by systemic inflammation. Interleukin-6 signaling and cyclooxygenase-1 mediated prostaglandin synthesis in the microglia were critical for the inflammation-induced aversion. Correspondingly, microglial activation led to a prostaglandin-dependent reduction of the excitability of striatal neurons. These findings demonstrate a mechanism by which microglial activation causes negative affect through prostaglandin-dependent modulation of striatal neurons and indicate that interference with this mechanism could milden the depressive symptoms in somatic and psychiatric diseases involving microglial activation.


Assuntos
Anedonia/fisiologia , Corpo Estriado/imunologia , Depressão/imunologia , Microglia/imunologia , Neurônios/fisiologia , Animais , Animais Geneticamente Modificados , Comportamento Animal , Células Cultivadas , Modelos Animais de Doenças , Humanos , Inflamação , Interleucina-6/metabolismo , Ativação de Macrófagos , Camundongos , Inflamação Neurogênica , Prostaglandinas/metabolismo
11.
CA Cancer J Clin ; 72(5): 490-502, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35709081

RESUMO

Depression is highly prevalent in those diagnosed with cancer and is also associated with poorer prognostic outcomes. Mindfulness-based interventions are effective in reducing depressive symptoms and improving quality of life in patients with cancer. The objective of this review was to investigate whether mindfulness practices can improve survival and, if so, what mechanisms of action may contribute to these outcomes. Although no long-term studies have investigated this hypothesis, the current literature supports an inflammatory basis for depression, implicating proinflammatory cytokines and hypothalamic-pituitary-adrenal axis dysfunction as contributing factors. Markers of inflammation, such as interleukin-6, tumor necrosis factor-α, and cortisol, are all found at elevated concentrations in many depressed individuals. These exact mechanisms are associated with higher mortality in patients with cancer. Mindfulness has been studied for its effects on cytokine and cortisol levels, and there are promising data to support that the intervention can measurably decrease inflammation. Therefore, it is conceivable that mindfulness programs can affect survival in this population. There are limited data on the long-term effects of mindfulness on depression and inflammatory markers in patients with cancer, and there are potential barriers to the implementation of mindfulness-based interventions as part of a comprehensive treatment plan. Therefore, it is necessary to further explore these questions through longitudinal studies to establish a survival correlation. CA Cancer J Clin. 2022;72:490-502.


Assuntos
Atenção Plena , Neoplasias , Depressão/terapia , Humanos , Hidrocortisona , Sistema Hipotálamo-Hipofisário , Inflamação/terapia , Neoplasias/terapia , Sistema Hipófise-Suprarrenal , Qualidade de Vida
12.
Cell ; 159(1): 33-45, 2014 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-25259918

RESUMO

Depression is a debilitating condition with a profound impact on quality of life for millions of people worldwide. Physical exercise is used as a treatment strategy for many patients, but the mechanisms that underlie its beneficial effects remain unknown. Here, we describe a mechanism by which skeletal muscle PGC-1α1 induced by exercise training changes kynurenine metabolism and protects from stress-induced depression. Activation of the PGC-1α1-PPARα/δ pathway increases skeletal muscle expression of kynurenine aminotransferases, thus enhancing the conversion of kynurenine into kynurenic acid, a metabolite unable to cross the blood-brain barrier. Reducing plasma kynurenine protects the brain from stress-induced changes associated with depression and renders skeletal muscle-specific PGC-1α1 transgenic mice resistant to depression induced by chronic mild stress or direct kynurenine administration. This study opens therapeutic avenues for the treatment of depression by targeting the PGC-1α1-PPAR axis in skeletal muscle, without the need to cross the blood-brain barrier.


Assuntos
Depressão/prevenção & controle , Cinurenina/metabolismo , Músculo Esquelético/enzimologia , Estresse Psicológico/complicações , Fatores de Transcrição/metabolismo , Animais , Barreira Hematoencefálica , Depressão/metabolismo , Perfilação da Expressão Gênica , Humanos , Ácido Cinurênico , Camundongos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , PPAR alfa/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Condicionamento Físico Animal , Condicionamento Físico Humano , Transaminases/metabolismo , Fatores de Transcrição/genética
13.
Nature ; 622(7981): 130-138, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37730990

RESUMO

Deep brain stimulation (DBS) of the subcallosal cingulate (SCC) can provide long-term symptom relief for treatment-resistant depression (TRD)1. However, achieving stable recovery is unpredictable2, typically requiring trial-and-error stimulation adjustments due to individual recovery trajectories and subjective symptom reporting3. We currently lack objective brain-based biomarkers to guide clinical decisions by distinguishing natural transient mood fluctuations from situations requiring intervention. To address this gap, we used a new device enabling electrophysiology recording to deliver SCC DBS to ten TRD participants (ClinicalTrials.gov identifier NCT01984710). At the study endpoint of 24 weeks, 90% of participants demonstrated robust clinical response, and 70% achieved remission. Using SCC local field potentials available from six participants, we deployed an explainable artificial intelligence approach to identify SCC local field potential changes indicating the patient's current clinical state. This biomarker is distinct from transient stimulation effects, sensitive to therapeutic adjustments and accurate at capturing individual recovery states. Variable recovery trajectories are predicted by the degree of preoperative damage to the structural integrity and functional connectivity within the targeted white matter treatment network, and are matched by objective facial expression changes detected using data-driven video analysis. Our results demonstrate the utility of objective biomarkers in the management of personalized SCC DBS and provide new insight into the relationship between multifaceted (functional, anatomical and behavioural) features of TRD pathology, motivating further research into causes of variability in depression treatment.


Assuntos
Estimulação Encefálica Profunda , Depressão , Transtorno Depressivo Maior , Humanos , Inteligência Artificial , Biomarcadores , Estimulação Encefálica Profunda/métodos , Depressão/fisiopatologia , Depressão/terapia , Transtorno Depressivo Maior/fisiopatologia , Transtorno Depressivo Maior/terapia , Eletrofisiologia , Resultado do Tratamento , Medida de Potenciais de Campo Local , Substância Branca , Lobo Límbico/fisiologia , Lobo Límbico/fisiopatologia , Expressão Facial
14.
Nature ; 622(7984): 802-809, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37853123

RESUMO

Ketamine, an N-methyl-D-aspartate receptor (NMDAR) antagonist1, has revolutionized the treatment of depression because of its potent, rapid and sustained antidepressant effects2-4. Although the elimination half-life of ketamine is only 13 min in mice5, its antidepressant activities can last for at least 24 h6-9. This large discrepancy poses an interesting basic biological question and has strong clinical implications. Here we demonstrate that after a single systemic injection, ketamine continues to suppress burst firing and block NMDARs in the lateral habenula (LHb) for up to 24 h. This long inhibition of NMDARs is not due to endocytosis but depends on the use-dependent trapping of ketamine in NMDARs. The rate of untrapping is regulated by neural activity. Harnessing the dynamic equilibrium of ketamine-NMDAR interactions by activating the LHb and opening local NMDARs at different plasma ketamine concentrations, we were able to either shorten or prolong the antidepressant effects of ketamine in vivo. These results provide new insights into the causal mechanisms of the sustained antidepressant effects of ketamine. The ability to modulate the duration of ketamine action based on the biophysical properties of ketamine-NMDAR interactions opens up new opportunities for the therapeutic use of ketamine.


Assuntos
Antidepressivos , Depressão , Habenula , Ketamina , Receptores de N-Metil-D-Aspartato , Animais , Camundongos , Antidepressivos/administração & dosagem , Antidepressivos/metabolismo , Antidepressivos/farmacocinética , Antidepressivos/farmacologia , Depressão/tratamento farmacológico , Depressão/metabolismo , Habenula/efeitos dos fármacos , Habenula/metabolismo , Meia-Vida , Ketamina/administração & dosagem , Ketamina/metabolismo , Ketamina/farmacocinética , Ketamina/farmacologia , Neurônios/fisiologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Fatores de Tempo , Ligação Proteica
15.
Annu Rev Neurosci ; 43: 355-374, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32109184

RESUMO

Opioid addiction and overdose are at record levels in the United States. This is driven, in part, by their widespread prescription for the treatment of pain, which also increased opportunity for diversion by sensation-seeking users. Despite considerable research on the neurobiology of addiction, treatment options for opioid abuse remain limited. Mood disorders, particularly depression, are often comorbid with both pain disorders and opioid abuse. The endogenous opioid system, a complex neuromodulatory system, sits at the neurobiological convergence point of these three comorbid disease states. We review evidence for dysregulation of the endogenous opioid system as a mechanism for the development of opioid addiction and/or mood disorder. Specifically, individual differences in opioid system function may underlie differences in vulnerability to opioid addiction and mood disorders. We also review novel research, which promises to provide more detailed understanding of individual differences in endogenous opioid neurobiology and its contribution to opioid addiction susceptibility.


Assuntos
Analgésicos Opioides/uso terapêutico , Dor Crônica/tratamento farmacológico , Depressão/tratamento farmacológico , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Animais , Overdose de Drogas/tratamento farmacológico , Humanos , Medicina de Precisão/métodos
16.
PLoS Biol ; 22(2): e3002518, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38386616

RESUMO

Neurons in the subthalamic nucleus (STN) become hyperactive following nerve injury and promote pain-related responses in mice. Considering that the anterior cingulate cortex (ACC) is involved in pain and emotion processing and projects to the STN, we hypothesize that ACC neurons may contribute to hyperactivity in STN neurons in chronic pain. In the present study, we showed that ACC neurons enhanced activity in response to noxious stimuli and to alterations in emotional states and became hyperactive in chronic pain state established by spared nerve injury of the sciatic nerve (SNI) in mice. In naïve mice, STN neurons were activated by noxious stimuli, but not by alterations in emotional states. Pain responses in STN neurons were attenuated in both naïve and SNI mice when ACC neurons were inhibited. Furthermore, optogenetic activation of the ACC-STN pathway induced bilateral hyperalgesia and depression-like behaviors in naive mice; conversely, inhibition of this pathway is sufficient to attenuate hyperalgesia and depression-like behaviors in SNI mice and naïve mice subjected to stimulation of STN neurons. Finally, mitigation of pain-like and depression-like behaviors in SNI mice by inhibition of the ACC-STN projection was eliminated by activation of STN neurons. Our results demonstrate that hyperactivity in the ACC-STN pathway may be an important pathophysiology in comorbid chronic pain and depression. Thus, the ACC-STN pathway may be an intervention target for the treatment of the comorbid chronic pain and depression.


Assuntos
Dor Crônica , Camundongos , Masculino , Animais , Giro do Cíngulo/fisiologia , Hiperalgesia , Depressão , Neurônios/fisiologia
17.
Cell ; 149(5): 1152-63, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22632977

RESUMO

Our understanding of current treatments for depression, and the development of more specific therapies, is limited by the complexity of the circuits controlling mood and the distributed actions of antidepressants. Although the therapeutic efficacy of serotonin-specific reuptake inhibitors (SSRIs) is correlated with increases in cortical activity, the cell types crucial for their action remain unknown. Here we employ bacTRAP translational profiling to show that layer 5 corticostriatal pyramidal cells expressing p11 (S100a10) are strongly and specifically responsive to chronic antidepressant treatment. This response requires p11 and includes the specific induction of Htr4 expression. Cortex-specific deletion of p11 abolishes behavioral responses to SSRIs, but does not lead to increased depression-like behaviors. Our data identify corticostriatal projection neurons as critical for the response to antidepressants, and suggest that the regulation of serotonergic tone in this single cell type plays a pivotal role in antidepressant therapy.


Assuntos
Antidepressivos/metabolismo , Depressão/tratamento farmacológico , Neurônios/citologia , Córtex Pré-Frontal/citologia , Inibidores Seletivos de Recaptação de Serotonina/metabolismo , Animais , Antidepressivos/farmacologia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutação , Córtex Pré-Frontal/metabolismo , Células Piramidais/metabolismo , Proteínas S100/genética , Proteínas S100/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia
18.
Nature ; 589(7842): 474-479, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33299186

RESUMO

The psychedelic alkaloid ibogaine has anti-addictive properties in both humans and animals1. Unlike most medications for the treatment of substance use disorders, anecdotal reports suggest that ibogaine has the potential to treat addiction to various substances, including opiates, alcohol and psychostimulants. The effects of ibogaine-like those of other psychedelic compounds-are long-lasting2, which has been attributed to its ability to modify addiction-related neural circuitry through the activation of neurotrophic factor signalling3,4. However, several safety concerns have hindered the clinical development of ibogaine, including its toxicity, hallucinogenic potential and tendency to induce cardiac arrhythmias. Here we apply the principles of function-oriented synthesis to identify the key structural elements of the potential therapeutic pharmacophore of ibogaine, and we use this information to engineer tabernanthalog-a water-soluble, non-hallucinogenic, non-toxic analogue of ibogaine that can be prepared in a single step. In rodents, tabernanthalog was found to promote structural neural plasticity, reduce alcohol- and heroin-seeking behaviour, and produce antidepressant-like effects. This work demonstrates that, through careful chemical design, it is possible to modify a psychedelic compound to produce a safer, non-hallucinogenic variant that has therapeutic potential.


Assuntos
Comportamento Aditivo/tratamento farmacológico , Desenho de Fármacos , Ibogaína/análogos & derivados , Ibogaína/efeitos adversos , Alcoolismo/tratamento farmacológico , Animais , Antidepressivos/farmacologia , Arritmias Cardíacas/induzido quimicamente , Técnicas de Química Sintética , Depressão/tratamento farmacológico , Modelos Animais de Doenças , Feminino , Alucinógenos/efeitos adversos , Dependência de Heroína/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasticidade Neuronal/efeitos dos fármacos , Segurança do Paciente , Receptor 5-HT2A de Serotonina/metabolismo , Agonistas do Receptor 5-HT2 de Serotonina/farmacologia , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico , Natação , Tabernaemontana/química
19.
Proc Natl Acad Sci U S A ; 121(14): e2319837121, 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38530887

RESUMO

Depression has robust natural language correlates and can increasingly be measured in language using predictive models. However, despite evidence that language use varies as a function of individual demographic features (e.g., age, gender), previous work has not systematically examined whether and how depression's association with language varies by race. We examine how race moderates the relationship between language features (i.e., first-person pronouns and negative emotions) from social media posts and self-reported depression, in a matched sample of Black and White English speakers in the United States. Our findings reveal moderating effects of race: While depression severity predicts I-usage in White individuals, it does not in Black individuals. White individuals use more belongingness and self-deprecation-related negative emotions. Machine learning models trained on similar amounts of data to predict depression severity performed poorly when tested on Black individuals, even when they were trained exclusively using the language of Black individuals. In contrast, analogous models tested on White individuals performed relatively well. Our study reveals surprising race-based differences in the expression of depression in natural language and highlights the need to understand these effects better, especially before language-based models for detecting psychological phenomena are integrated into clinical practice.


Assuntos
Depressão , Mídias Sociais , Humanos , Estados Unidos , Depressão/psicologia , Emoções , Idioma
20.
Proc Natl Acad Sci U S A ; 121(21): e2319595121, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38739786

RESUMO

As a global problem, fine particulate matter (PM2.5) really needs local fixes. Considering the increasing epidemiological relevance to anxiety and depression but inconsistent toxicological results, the most important question is to clarify whether and how PM2.5 causally contributes to these mental disorders and which components are the most dangerous for crucial mitigation in a particular place. In the present study, we chronically subjected male mice to a real-world PM2.5 exposure system throughout the winter heating period in a coal combustion area and revealed that PM2.5 caused anxiety and depression-like behaviors in adults such as restricted activity, diminished exploratory interest, enhanced repetitive stereotypy, and elevated acquired immobility, through behavioral tests including open field, elevated plus maze, marble-burying, and forced swimming tests. Importantly, we found that dopamine signaling was perturbed using mRNA transcriptional profile and bioinformatics analysis, with Drd1 as a potential target. Subsequently, we developed the Drd1 expression-directed multifraction isolating and nontarget identifying framework and identified a total of 209 compounds in PM2.5 organic extracts capable of reducing Drd1 expression. Furthermore, by applying hierarchical characteristic fragment analysis and molecular docking and dynamics simulation, we clarified that phenyl-containing compounds competitively bound to DRD1 and interfered with dopamine signaling, thereby contributing to mental disorders. Taken together, this work provides experimental evidence for researchers and clinicians to identify hazardous factors in PM2.5 and prevent adverse health outcomes and for local governments and municipalities to control source emissions for diminishing specific disease burdens.


Assuntos
Ansiedade , Depressão , Material Particulado , Receptores de Dopamina D1 , Animais , Material Particulado/toxicidade , Camundongos , Masculino , Ansiedade/metabolismo , Depressão/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D1/genética , Poluentes Atmosféricos/toxicidade , Comportamento Animal/efeitos dos fármacos , Simulação de Acoplamento Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA