1
|
Fergusson KN, Tanner JL, Brand JA, Hannington SL, Pettersen AK, Sundin J, Saaristo M, Bertram MG, Martin JM, Wong BBM. Effects of long-term fluoxetine exposure on morphology, but not behaviour or metabolic rate, in male guppies (Poecilia reticulata). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 276:107082. [PMID: 39270523 DOI: 10.1016/j.aquatox.2024.107082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024]
Abstract
Contamination of aquatic ecosystems by pharmaceuticals is a growing threat worldwide. The antidepressant fluoxetine is one such pharmaceutical that is frequently detected in aquatic ecosystems, and has been found to alter the behaviour and physiology of exposed wildlife. Few studies, however, have investigated potential combined effects on behaviour and metabolic rate. In addition, exposures are often short in duration and rarely conducted under ecologically relevant conditions. Here, we examined the impacts of long-term fluoxetine exposure on boldness (exploration, activity, and antipredator behaviour), metabolic rate, and morphology in male guppies (Poecilia reticulata). Specifically, fish were exposed for 8 months (corresponding to approximately two overlapping generations) in semi-natural mesocosms to one of three treatments: an unexposed control (0 ng L-1), or low or high fluoxetine (mean measured concentrations: 30 ng L-1 and 292 ng L-1, respectively). Following exposure, we quantified male exploratory behaviour and activity in a novel environment (maze arena) and antipredator behaviour in the presence or absence of a live predator (spangled perch, Leiopotherapon unicolor), as well as metabolic rate and morphology (mass, standard length, and scaled mass index). Fluoxetine exposure did not significantly alter boldness, metabolic rate, mass, or standard length. However, fluoxetine exposure did alter body condition, whereby fish in the high treatment had a higher scaled mass index than control fish. Our results, considered alongside previous work, underscore the importance of exposure duration in mediating the effects of fluoxetine on fitness-related traits. Continued research under extended exposure periods (i.e., spanning multiple generations) is essential if we are to accurately predict the ecological impacts of fluoxetine on exposed wildlife, and their underlying mechanism(s).
Collapse
Affiliation(s)
- Kate N Fergusson
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia.
| | - James L Tanner
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia.
| | - Jack A Brand
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia; Department of Wildlife, Fish, and Environmental Studies, Swedish University of Agricultural Sciences, Umeå, Sweden.
| | | | - Amanda K Pettersen
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia; School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia.
| | - Josefin Sundin
- Department of Aquatic Resources, Swedish University of Agricultural Sciences, Drottningholm, Sweden.
| | - Minna Saaristo
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia; Environment Protection Authority Victoria, EPA Science, Macleod, Victoria, Australia.
| | - Michael G Bertram
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia; Department of Wildlife, Fish, and Environmental Studies, Swedish University of Agricultural Sciences, Umeå, Sweden; Department of Zoology, Stockholm University, Stockholm, Sweden.
| | - Jake M Martin
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia; Department of Wildlife, Fish, and Environmental Studies, Swedish University of Agricultural Sciences, Umeå, Sweden; Department of Zoology, Stockholm University, Stockholm, Sweden; School of Life and Environmental Sciences, Deakin University, Waurn Ponds, Geelong, Australia.
| | - Bob B M Wong
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
2
|
Fujii C, Zorumski CF, Izumi Y. Endoplasmic reticulum stress, autophagy, neuroinflammation, and sigma 1 receptors as contributors to depression and its treatment. Neural Regen Res 2024; 19:2202-2211. [PMID: 38488553 PMCID: PMC11034583 DOI: 10.4103/1673-5374.391334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/02/2023] [Accepted: 11/24/2023] [Indexed: 04/24/2024] Open
Abstract
The etiological factors contributing to depression and other neuropsychiatric disorders are largely undefined. Endoplasmic reticulum stress pathways and autophagy are well-defined mechanisms that play critical functions in recognizing and resolving cellular stress and are possible targets for the pathophysiology and treatment of psychiatric and neurologic illnesses. An increasing number of studies indicate the involvement of endoplasmic reticulum stress and autophagy in the control of neuroinflammation, a contributing factor to multiple neuropsychiatric illnesses. Initial inflammatory triggers induce endoplasmic reticulum stress, leading to neuroinflammatory responses. Subsequently, induction of autophagy by neurosteroids and other signaling pathways that converge on autophagy induction are thought to participate in resolving neuroinflammation. The aim of this review is to summarize our current understanding of the molecular mechanisms governing the induction of endoplasmic reticulum stress, autophagy, and neuroinflammation in the central nervous system. Studies focused on innate immune factors, including neurosteroids with anti-inflammatory roles will be reviewed. In the context of depression, animal models that led to our current understanding of molecular mechanisms underlying depression will be highlighted, including the roles of sigma 1 receptors and pharmacological agents that dampen endoplasmic reticulum stress and associated neuroinflammation.
Collapse
Affiliation(s)
- Chika Fujii
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Charles F. Zorumski
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Yukitoshi Izumi
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
3
|
Bremshey S, Groß J, Renken K, Masseck OA. The role of serotonin in depression-A historical roundup and future directions. J Neurochem 2024; 168:1751-1779. [PMID: 38477031 DOI: 10.1111/jnc.16097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024]
Abstract
Depression is one of the most common psychiatric disorders worldwide, affecting approximately 280 million people, with probably much higher unrecorded cases. Depression is associated with symptoms such as anhedonia, feelings of hopelessness, sleep disturbances, and even suicidal thoughts. Tragically, more than 700 000 people commit suicide each year. Although depression has been studied for many decades, the exact mechanisms that lead to depression are still unknown, and available treatments only help a fraction of patients. In the late 1960s, the serotonin hypothesis was published, suggesting that serotonin is the key player in depressive disorders. However, this hypothesis is being increasingly doubted as there is evidence for the influence of other neurotransmitters, such as noradrenaline, glutamate, and dopamine, as well as larger systemic causes such as altered activity in the limbic network or inflammatory processes. In this narrative review, we aim to contribute to the ongoing debate on the involvement of serotonin in depression. We will review the evolution of antidepressant treatments, systemic research on depression over the years, and future research applications that will help to bridge the gap between systemic research and neurotransmitter dynamics using biosensors. These new tools in combination with systemic applications, will in the future provide a deeper understanding of the serotonergic dynamics in depression.
Collapse
Affiliation(s)
- Svenja Bremshey
- Synthetic Biology, University of Bremen, Bremen, Germany
- Neuropharmacology, University of Bremen, Bremen, Germany
| | - Juliana Groß
- Synthetic Biology, University of Bremen, Bremen, Germany
| | - Kim Renken
- Synthetic Biology, University of Bremen, Bremen, Germany
| | | |
Collapse
|
4
|
Lu Y, Wang S, Shen Y. Theoretical insights of the pharmaceutical compound fluoxetine in water: Role in direct photolysis and indirect photolysis by free radicals. J Environ Sci (China) 2024; 142:269-278. [PMID: 38527892 DOI: 10.1016/j.jes.2023.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/03/2023] [Accepted: 07/18/2023] [Indexed: 03/27/2024]
Abstract
The frequent detection of pharmaceutical compounds in the environment has led to a growing awareness, which may pose a major threat to the aquatic environment. In this study, photodegradation (direct and indirect photolysis) of two different dissociation states of fluoxetine (FLU) was investigated in water, mainly including the determination of photolytic transition states and products, and the mechanisms of indirect photodegradation with ·OH, CO3*- and NO3*. The main direct photolysis pathways are defluorination and C-C bond cleavage. In addition, the indirect photodegradation of FLU in water is mainly through the reactions with ·OH and NO3*, and the photodegradation reaction with CO3*- is relatively difficult to occur in the water environment. Our results provide a theoretical basis for understanding the phototransformation process of FLU in the water environment and assessing its potential risk.
Collapse
Affiliation(s)
- Ying Lu
- School of Environmental Science and Engineering, Collaborative Innovation Center of Atmospheric Environment and Equipment Technology, Jiangsu Key Laboratory of Atmospheric Environment Monitoring and Pollution Control, Nanjing University of Information Science and Technology, Nanjing 210044, China
| | - Se Wang
- School of Environmental Science and Engineering, Collaborative Innovation Center of Atmospheric Environment and Equipment Technology, Jiangsu Key Laboratory of Atmospheric Environment Monitoring and Pollution Control, Nanjing University of Information Science and Technology, Nanjing 210044, China.
| | - Yifan Shen
- School of Environmental Science and Engineering, Collaborative Innovation Center of Atmospheric Environment and Equipment Technology, Jiangsu Key Laboratory of Atmospheric Environment Monitoring and Pollution Control, Nanjing University of Information Science and Technology, Nanjing 210044, China
| |
Collapse
|
5
|
Fefeu M, Blatzer M, Kneppers A, Briand D, Rocheteau P, Haroche A, Hardy D, Juchet-Martin M, Danckaert A, Coudoré F, Tutakhail A, Huchet C, Lafoux A, Mounier R, Mir O, Gaillard R, Chrétien F. Serotonin reuptake inhibitors improve muscle stem cell function and muscle regeneration in male mice. Nat Commun 2024; 15:6457. [PMID: 39085209 PMCID: PMC11291725 DOI: 10.1038/s41467-024-50220-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 07/03/2024] [Indexed: 08/02/2024] Open
Abstract
Serotonin reuptake inhibitor antidepressants such as fluoxetine are widely used to treat mood disorders. The mechanisms of action include an increase in extracellular level of serotonin, neurogenesis, and growth of vessels in the brain. We investigated whether fluoxetine could have broader peripheral regenerative properties. Following prolonged administration of fluoxetine in male mice, we showed that fluoxetine increases the number of muscle stem cells and muscle angiogenesis, associated with positive changes in skeletal muscle function. Fluoxetine also improved skeletal muscle regeneration after single and multiples injuries with an increased muscle stem cells pool and vessel density associated with reduced fibrotic lesions and inflammation. Mice devoid of peripheral serotonin treated with fluoxetine did not exhibit beneficial effects during muscle regeneration. Specifically, pharmacological, and genetic inactivation of the 5-HT1B subtype serotonin receptor also abolished the enhanced regenerative process induced by fluoxetine. We highlight here a regenerative property of serotonin on skeletal muscle.
Collapse
Affiliation(s)
- Mylène Fefeu
- GHU Paris Psychiatrie & Neurosciences, site Sainte Anne, Service Hospitalo-Universitaire de psychiatrie, Paris, France
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France
- Université de Paris Cité, Paris, France
| | - Michael Blatzer
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France
| | - Anita Kneppers
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - David Briand
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France
| | - Pierre Rocheteau
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France
| | - Alexandre Haroche
- GHU Paris Psychiatrie & Neurosciences, site Sainte Anne, Service Hospitalo-Universitaire de psychiatrie, Paris, France
| | - David Hardy
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France
| | - Mélanie Juchet-Martin
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France
| | | | - François Coudoré
- CESP, MOODS Team, Inserm, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France
| | - Abdulkarim Tutakhail
- CESP, MOODS Team, Inserm, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France
| | - Corinne Huchet
- TaRGeT, INSERM UMR 1089, Nantes Université, CHU Nantes, Nantes, France
| | - Aude Lafoux
- Therassay Platform, Capacités, Université de Nantes, IRS 2 Nantes Biotech, Nantes, France
| | - Rémi Mounier
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Olivier Mir
- Sarcoma Group, Gustave Roussy, Villejuif, France
| | - Raphaël Gaillard
- GHU Paris Psychiatrie & Neurosciences, site Sainte Anne, Service Hospitalo-Universitaire de psychiatrie, Paris, France.
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France.
- Université de Paris Cité, Paris, France.
| | - Fabrice Chrétien
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France.
- Université de Paris Cité, Paris, France.
- GHU Paris Psychiatrie & Neurosciences, site Sainte Anne, Service Hospitalo-Universitaire de neuropathologie, Paris, France.
| |
Collapse
|
6
|
Ampuero E, Luarte A, Flores FS, Soto AI, Pino C, Silva V, Erlandsen M, Concha T, Wyneken U. The multifaceted effects of fluoxetine treatment on cognitive functions. Front Pharmacol 2024; 15:1412420. [PMID: 39081952 PMCID: PMC11286485 DOI: 10.3389/fphar.2024.1412420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/10/2024] [Indexed: 08/02/2024] Open
Abstract
Fluoxetine, the prototypical selective serotonin reuptake inhibitor (SSRI), is widely used to treat major depressive disorder (MDD) and a variety of other central nervous system conditions, primarily due to its established clinical safety profile. Although its efficacy in treating depression is well-recognized, the impact of fluoxetine on cognitive functions remains inconsistent and elusive. In this review, we first examine the well-substantiated biological mechanisms underlying fluoxetine's antidepressant effects, which include serotonin reuptake inhibition and activation of TrkB receptors-key to brain-derived neurotrophic factor (BDNF) signaling. Subsequently, we delve into the cognitive side effects observed in both preclinical and clinical studies, affecting domains such as memory, attention, and executive functions. While certain studies indicate cognitive improvements in patients with underlying disorders, there is also evidence of negative effects, influenced by variables like gender, duration of treatment, age, disease pathology, and the specifics of cognitive testing. Significantly, the negative cognitive outcomes reported in preclinical research often involve healthy, non-diseased animals. This review underscores the necessity for heightened caution in fluoxetine prescription and further investigation into its potentially detrimental cognitive effects, even when used prophylactically.
Collapse
Affiliation(s)
- Estíbaliz Ampuero
- Laboratorio Neurofarmacología del Comportamiento, Facultad de Química y Biología, Universidad de Santiago, Santiago, Chile
| | - Alejandro Luarte
- Laboratorio Neurociencias, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Francisca Sofia Flores
- Laboratorio Neurociencias, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Antonia Ignacia Soto
- Laboratorio Neurociencias, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Catalina Pino
- Laboratorio Neurociencias, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Viviana Silva
- Laboratorio Neurociencias, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Macarena Erlandsen
- Laboratorio Neurociencias, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Teresita Concha
- Laboratorio Neurociencias, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Ursula Wyneken
- Laboratorio Neurociencias, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| |
Collapse
|
7
|
Lu X, Wang X, Chen S, Fan T, Zhao L, Zhong R, Sun G. The rat acute oral toxicity of trifluoromethyl compounds (TFMs): a computational toxicology study combining the 2D-QSTR, read-across and consensus modeling methods. Arch Toxicol 2024; 98:2213-2229. [PMID: 38627326 DOI: 10.1007/s00204-024-03739-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/18/2024] [Indexed: 06/13/2024]
Abstract
All areas of the modern society are affected by fluorine chemistry. In particular, fluorine plays an important role in medical, pharmaceutical and agrochemical sciences. Amongst various fluoro-organic compounds, trifluoromethyl (CF3) group is valuable in applications such as pharmaceuticals, agrochemicals and industrial chemicals. In the present study, following the strict OECD modelling principles, a quantitative structure-toxicity relationship (QSTR) modelling for the rat acute oral toxicity of trifluoromethyl compounds (TFMs) was established by genetic algorithm-multiple linear regression (GA-MLR) approach. All developed models were evaluated by various state-of-the-art validation metrics and the OECD principles. The best QSTR model included nine easily interpretable 2D molecular descriptors with clear physical and chemical significance. The mechanistic interpretation showed that the atom-type electro-topological state indices, molecular connectivity, ionization potential, lipophilicity and some autocorrelation coefficients are the main factors contributing to the acute oral toxicity of TFMs against rats. To validate that the selected 2D descriptors can effectively characterize the toxicity, we performed the chemical read-across analysis. We also compared the best QSTR model with public OPERA tool to demonstrate the reliability of the predictions. To further improve the prediction range of the QSTR model, we performed the consensus modelling. Finally, the optimum QSTR model was utilized to predict a true external set containing many untested/unknown TFMs for the first time. Overall, the developed model contributes to a more comprehensive safety assessment approach for novel CF3-containing pharmaceuticals or chemicals, reducing unnecessary chemical synthesis whilst saving the development cost of new drugs.
Collapse
Affiliation(s)
- Xinyi Lu
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, People's Republic of China
| | - Xin Wang
- Department of Clinical Trials Center, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, People's Republic of China
| | - Shuo Chen
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, People's Republic of China
| | - Tengjiao Fan
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, People's Republic of China
- Department of Medical Technology, Beijing Pharmaceutical University of Staff and Workers, Beijing, 100079, China
| | - Lijiao Zhao
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, People's Republic of China
| | - Rugang Zhong
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, People's Republic of China
| | - Guohui Sun
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, People's Republic of China.
| |
Collapse
|
8
|
Seki T, Katsura M, Yamasaki M, Yamashita K, Kokushi E, Uno S. Effects of diphenhydramine exposure on reproduction of mature Japanese medaka (Oryzias latipes). CHEMOSPHERE 2024; 358:142163. [PMID: 38697572 DOI: 10.1016/j.chemosphere.2024.142163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/16/2024] [Accepted: 04/25/2024] [Indexed: 05/05/2024]
Abstract
Diphenhydramine (DPH) is an antihistamine drug. It has been frequently detected in the environment, because it is not completely degraded in wastewater treatment plants. Recent studies have shown the adverse effects of DPH exposure to various aquatic organisms; however, its chronic effects on fish have been poorly elucidated. In this study, several pairs of mature Japanese medaka (Oryzias latipes) were exposed to DPH for a long period to determine the effects of DPH exposure on the subsequent generations, number of spawned and fertilized eggs, expression of sex-related genes, feeding behavior, embryo development, hatching rate, malformations among the hatched larvae, and mortality rate. The number of spawned eggs significantly decreased, when the parent fish were continuously exposed to 31.6 μg/L DPH for over 46 days. DPH exposure also altered the feeding behavior of medaka individuals, and increased the larval mortality rate. The effects of DPH exposure to fish may occur to some extent in the actual aquatic environment, although the risk evaluations in the field are limited.
Collapse
Affiliation(s)
- Tamaki Seki
- The United Graduate School of Agricultural Science, Kagoshima University, 50-20 Shimoarata 4-Chome, Kagoshima, 890-0056, Japan
| | - Motoaki Katsura
- Education and Research Center for Marine Resources and Environment, Faculty of Fisheries, Kagoshima University, 50-20 Shimoarata 4-Chome, Kagoshima, 890-0056, Japan
| | - Masatoshi Yamasaki
- Education and Research Center for Marine Resources and Environment, Faculty of Fisheries, Kagoshima University, 50-20 Shimoarata 4-Chome, Kagoshima, 890-0056, Japan
| | - Kazuki Yamashita
- Education and Research Center for Marine Resources and Environment, Faculty of Fisheries, Kagoshima University, 50-20 Shimoarata 4-Chome, Kagoshima, 890-0056, Japan
| | - Emiko Kokushi
- Education and Research Center for Marine Resources and Environment, Faculty of Fisheries, Kagoshima University, 50-20 Shimoarata 4-Chome, Kagoshima, 890-0056, Japan
| | - Seiichi Uno
- Education and Research Center for Marine Resources and Environment, Faculty of Fisheries, Kagoshima University, 50-20 Shimoarata 4-Chome, Kagoshima, 890-0056, Japan.
| |
Collapse
|
9
|
Morán-Serradilla C, Plano D, Sanmartín C, Sharma AK. Selenization of Small Molecule Drugs: A New Player on the Board. J Med Chem 2024; 67:7759-7787. [PMID: 38716896 DOI: 10.1021/acs.jmedchem.3c02426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2024]
Abstract
There is an urgent need to develop safer and more effective modalities for the treatment of a wide range of pathologies due to the increasing rates of drug resistance, undesired side effects, poor clinical outcomes, etc. Throughout the years, selenium (Se) has attracted a great deal of attention due to its important role in human health. Besides, a growing body of work has unveiled that the inclusion of Se motifs into a great number of molecules is a promising strategy for obtaining novel therapeutic agents. In the current Perspective, we have gathered the most recent literature related to the incorporation of different Se moieties into the scaffolds of a wide range of known drugs and their feasible pharmaceutical applications. In addition, we highlight different representative examples as well as provide our perspective on Se drugs and the possible future directions, promises, opportunities, and challenges of this ground-breaking area of research.
Collapse
Affiliation(s)
| | - Daniel Plano
- Department of Pharmaceutical Sciences, University of Navarra, Irunlarrea 1, Pamplona E-31008, Spain
| | - Carmen Sanmartín
- Department of Pharmaceutical Sciences, University of Navarra, Irunlarrea 1, Pamplona E-31008, Spain
| | - Arun K Sharma
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey, Pennsylvania 17033, United States
- Penn State Cancer Institute, 400 University Drive,Hershey, Pennsylvania 17033, United States
| |
Collapse
|
10
|
Mir M, Khan AU, Khan A. Pharmacological investigation of taxifolin for its therapeutic potential in depression. Heliyon 2024; 10:e30467. [PMID: 38694040 PMCID: PMC11061746 DOI: 10.1016/j.heliyon.2024.e30467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/03/2024] Open
Abstract
The current study aimed to investigate the influence of taxifolin on depression symptoms alleviation in Male Sprague-Dawley rats by targeting underlying pathways of depression. Molecular docking analyses were conducted to validate taxifolin's binding affinities against various targets. In silico analysis of taxifolin revealed various aspects of post docking interactions with different protein targets. Depression was induced in rats via intraperitoneal injection of Lipopolysaccharide (LPS; 500 μ g/Kg) for 14 alternative days. Rats (n = 6/group) were randomly assigned to four groups: (i) Saline/Control, (ii) Disease (LPS 500 μg/kg), (iii) Standard (fluoxetine 20 mg/kg), and (iv) Treatment (taxifolin 20 mg/kg). At the end of the in vivo study, brain samples were used for biochemical and morphological analysis. Taxifolin exhibited neuroprotective effects, as evidenced by behavioral studies, antioxidant analysis, histopathological examination, immunohistochemistry, ELISA and RT PCR, indicating an increase number of surviving neurons, normalization of cell size and shape, and reduction in vacuolization. Taxifolin also decreased inflammatory markers such as TNF-α, NF-κb, IL-6 and COX-2, while significantly upregulating and activating the protective PPAR-γ pathway, through which it reduces the oxidative stress, neuroinflammation, neurodegeneration, thereby ameliorating depression symptoms in experimental rat model of depression. Our finding suggests that taxifolin act as neuroprotective agent partially mediated through PPAR-γ pathway.
Collapse
Affiliation(s)
- Maha Mir
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Arif-ullah Khan
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Aslam Khan
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| |
Collapse
|
11
|
Zhang W, Jin Y, Zhou FM. Chronic fluoxetine treatment desensitizes serotoninergic inhibition of GABA inputs and the intrinsic excitability of dorsal raphe serotonin neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.07.592963. [PMID: 38766100 PMCID: PMC11100661 DOI: 10.1101/2024.05.07.592963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Dorsal raphe serotonin (5-hydroxytryptamine, 5-HT) neurons are spontaneously active and release 5-HT that is critical to normal brain function such mood and emotion. Serotonin reuptake inhibitors (SSRIs) increase the synaptic and extracellular 5-HT level and are effective in treating depression. Treatment of two weeks or longer is often required for SSRIs to exert clinical benefits. The cellular mechanism underlying this delay was not fully understood. Here we show that the GABAergic inputs inhibit the spike firing of raphe 5-HT neurons; this GABAergic regulation was reduced by 5-HT, which was prevented by G-protein-activated inwardly rectifying potassium (Girk) channel inhibitor tertiapin-Q, indicating a contribution of 5-HT activation of Girk channels in GABAergic presynaptic axon terminals. Equally important, after 14 days of treatment of fluoxetine, a widely used SSRI type antidepressant, this 5-HT inhibition of GABAergic inputs was substantially downregulated. Furthermore, the chronic fluoxetine treatment substantially downregulated the 5-HT activation of the inhibitory Girk current in 5-HT neurons. Taken together, our results suggest that chronic fluoxetine administration, by blocking 5-HT reuptake and hence increasing the extracellular 5-HT level, can downregulate the function of 5-HT1B receptors on the GABAergic afferent axon terminals synapsing onto 5-HT neurons, allowing extrinsic, behaviorally important GABA neurons to more effectively influence 5-HT neurons; simultaneously, chronic fluoxetine treatment also downregulate somatic 5-HT autoreceptor-activated Girk channel-mediated hyperpolarization and decrease in input resistance and intrinsic excitability, rendering 5-HT neurons resistant to autoinhibition and leading to increased 5-HT neuron activity, potentially contributing to the antidepressant effect of SSRIs.
Collapse
|
12
|
Wang R, Hu X, Liu S, Wang J, Xiong F, Zhang X, Ye W, Wang H. Kaempferol-3-O-sophoroside (PCS-1) contributes to modulation of depressive-like behaviour in C57BL/6J mice by activating AMPK. Br J Pharmacol 2024; 181:1182-1202. [PMID: 37949672 DOI: 10.1111/bph.16283] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 10/18/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND AND PURPOSE Kaempferol-3-O-sophoroside (PCS-1) is the main component in Crocus sativus (Saffron), a herb with mood-enhancing properties. AMP-activated protein kinase (AMPK) is a potential therapeutic target for depression. This study explores the antidepressive-like properties of PCS-1 and its AMPK activation to confirm AMPK as a target for antidepression. EXPERIMENTAL APPROACH Corticosterone (CORT)-induced PC12 cell injury served as an in vitro model to evaluate the neuroprotective effect of PCS-1. Neuro-2a cells and primary neurons were utilized to evaluate the synaptogenesis role of PCS-1. CORT-induced mouse depression model and chronic unpredictable mild stress (CUMS) model were used to assess the antidepressive-like properties of PCS-1 through behavioural tests, magnetic resonance imaging, and biochemical index measurements. Western blot and immunofluorescence assays were used to study the mechanisms of PCS-1. Cellular thermal shift assay was used to confirm the binding target. KEY RESULTS PCS-1 (12.5-50 μM) ameliorated CORT-induced PC12 cell damage, oxidative stress and inflammation. PCS-1 alone promoted an increase in synapses in Neuro-2a cells and primary neurons. Oral administration of PCS-1 (10 and 20 mg·kg-1 ) ameliorated weight loss, dyskinesia, and hippocampal volume reduction induced by CORT and CUMS. PCS-1 bound to AMPK to improve the expression of brain-derived neurotrophic factor (BDNF) and induce autophagy. CONCLUSION AND IMPLICATIONS PCS-1 binds to AMPK to promote BDNF production and autophagy enhancement, ultimately achieving antidepressant effects. This study provides support for the clinical application of saffron petals and provides further evidence for AMPK as a potential target for antidepression.
Collapse
Affiliation(s)
- Rong Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Xiaolong Hu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Shumeng Liu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jingjin Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Fei Xiong
- State Key Laboratory of Bioelectronics, Jiangsu Laboratory for Biomaterials and Devices, Southeast University, Nanjing, People's Republic of China
| | - Xiaoqi Zhang
- Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou, People's Republic of China
| | - Wencai Ye
- Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou, People's Republic of China
| | - Hao Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
13
|
Su J, Yang L, Sun Z, Zhan X. Personalized Drug Therapy: Innovative Concept Guided With Proteoformics. Mol Cell Proteomics 2024; 23:100737. [PMID: 38354979 PMCID: PMC10950891 DOI: 10.1016/j.mcpro.2024.100737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/29/2024] [Accepted: 02/09/2024] [Indexed: 02/16/2024] Open
Abstract
Personalized medicine can reduce adverse effects, enhance drug efficacy, and optimize treatment outcomes, which represents the essence of personalized medicine in the pharmacy field. Protein drugs are crucial in the field of personalized drug therapy and are currently the mainstay, which possess higher target specificity and biological activity than small-molecule chemical drugs, making them efficient in regulating disease-related biological processes, and have significant potential in the development of personalized drugs. Currently, protein drugs are designed and developed for specific protein targets based on patient-specific protein data. However, due to the rapid development of two-dimensional gel electrophoresis and mass spectrometry, it is now widely recognized that a canonical protein actually includes multiple proteoforms, and the differences between these proteoforms will result in varying responses to drugs. The variation in the effects of different proteoforms can be significant and the impact can even alter the intended benefit of a drug, potentially making it harmful instead of lifesaving. As a result, we propose that protein drugs should shift from being targeted through the lens of protein (proteomics) to being targeted through the lens of proteoform (proteoformics). This will enable the development of personalized protein drugs that are better equipped to meet patients' specific needs and disease characteristics. With further development in the field of proteoformics, individualized drug therapy, especially personalized protein drugs aimed at proteoforms as a drug target, will improve the understanding of disease mechanisms, discovery of new drug targets and signaling pathways, provide a theoretical basis for the development of new drugs, aid doctors in conducting health risk assessments and making more cost-effective targeted prevention strategies conducted by artificial intelligence/machine learning, promote technological innovation, and provide more convenient treatment tailored to individualized patient profile, which will benefit the affected individuals and society at large.
Collapse
Affiliation(s)
- Junwen Su
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Lamei Yang
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ziran Sun
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xianquan Zhan
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
14
|
Yao H, Wang X, Chi J, Chen H, Liu Y, Yang J, Yu J, Ruan Y, Xiang X, Pi J, Xu JF. Exploring Novel Antidepressants Targeting G Protein-Coupled Receptors and Key Membrane Receptors Based on Molecular Structures. Molecules 2024; 29:964. [PMID: 38474476 DOI: 10.3390/molecules29050964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/29/2024] [Accepted: 02/09/2024] [Indexed: 03/14/2024] Open
Abstract
Major Depressive Disorder (MDD) is a complex mental disorder that involves alterations in signal transmission across multiple scales and structural abnormalities. The development of effective antidepressants (ADs) has been hindered by the dominance of monoamine hypothesis, resulting in slow progress. Traditional ADs have undesirable traits like delayed onset of action, limited efficacy, and severe side effects. Recently, two categories of fast-acting antidepressant compounds have surfaced, dissociative anesthetics S-ketamine and its metabolites, as well as psychedelics such as lysergic acid diethylamide (LSD). This has led to structural research and drug development of the receptors that they target. This review provides breakthroughs and achievements in the structure of depression-related receptors and novel ADs based on these. Cryo-electron microscopy (cryo-EM) has enabled researchers to identify the structures of membrane receptors, including the N-methyl-D-aspartate receptor (NMDAR) and the 5-hydroxytryptamine 2A (5-HT2A) receptor. These high-resolution structures can be used for the development of novel ADs using virtual drug screening (VDS). Moreover, the unique antidepressant effects of 5-HT1A receptors in various brain regions, and the pivotal roles of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) and tyrosine kinase receptor 2 (TrkB) in regulating synaptic plasticity, emphasize their potential as therapeutic targets. Using structural information, a series of highly selective ADs were designed based on the different role of receptors in MDD. These molecules have the favorable characteristics of rapid onset and low adverse drug reactions. This review offers researchers guidance and a methodological framework for the structure-based design of ADs.
Collapse
Affiliation(s)
- Hanbo Yao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Xiaodong Wang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Jiaxin Chi
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Haorong Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Yilin Liu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Jiayi Yang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Jiaqi Yu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Yongdui Ruan
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| | - Xufu Xiang
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jiang Pi
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Jun-Fa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
15
|
Lewis YD, Bergner L, Steinberg H, Bentley J, Himmerich H. Pharmacological Studies in Eating Disorders: A Historical Review. Nutrients 2024; 16:594. [PMID: 38474723 PMCID: PMC11154472 DOI: 10.3390/nu16050594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 03/14/2024] Open
Abstract
Eating disorders (EDs) are serious mental health conditions characterised by impaired eating behaviours and nutrition as well as disturbed body image, entailing considerable mortality and morbidity. Psychopharmacological medication is an important component in the treatment of EDs. In this review, we performed a historic analysis of pharmacotherapeutic research in EDs based on the scientific studies included in the recently published World Federation of Societies for Biological Psychiatry (WFSBP) guidelines for ED treatment. This analysis focuses on early approaches and trends in the methods of clinical pharmacological research in EDs, for example, the sample sizes of randomised controlled trials (RCTs). We found the development of psychopharmacological treatments for EDs followed advancements in psychiatric pharmacotherapy. However, the application of RCTs to the study of pharmacotherapy for EDs may be an impediment as limited participant numbers and inadequate research funding impede generalisability and statistical power. Moreover, current medication usage often deviates from guideline recommendations. In conclusion, the RCT model may not effectively capture the complexities of ED treatment, and funding limitations hinder research activity. Novel genetically/biologically based treatments are warranted. A more comprehensive understanding of EDs and individualised approaches should guide research and drug development for improved treatment outcomes.
Collapse
Affiliation(s)
- Yael D. Lewis
- Hadarim Eating Disorders Unit, Shalvata Mental Health Centre, Hod Hasharon 4534708, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Lukas Bergner
- Forschungsstelle für die Geschichte der Psychiatrie, Klinik und Poliklinik Psychiatrie und Psychotherapie, Medizinische Fakultät der Universität Leipzig, 04103 Leipzig, Germany; (L.B.); (H.S.)
| | - Holger Steinberg
- Forschungsstelle für die Geschichte der Psychiatrie, Klinik und Poliklinik Psychiatrie und Psychotherapie, Medizinische Fakultät der Universität Leipzig, 04103 Leipzig, Germany; (L.B.); (H.S.)
| | - Jessica Bentley
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London SE5 8AF, UK; (J.B.); (H.H.)
| | - Hubertus Himmerich
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London SE5 8AF, UK; (J.B.); (H.H.)
- South London and Maudsley NHS Foundation Trust, London BR3 3BX, UK
| |
Collapse
|
16
|
Abreu GR, Zaccarelli-Magalhães J, Féba LS, Penna EY, Silva JP, Teixeira MV, Manes M, Vergara GA, Rodrigues LMA, Fukushima AR, Ricci EL, Spinosa HS. Fluoxetine alters rat's milk properties causing impact on offspring's development. Toxicol Appl Pharmacol 2024; 483:116805. [PMID: 38191078 DOI: 10.1016/j.taap.2024.116805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/31/2023] [Accepted: 01/03/2024] [Indexed: 01/10/2024]
Abstract
Fluoxetine is an antidepressant used to treat several conditions including postpartum depression. This disease causes cognitive, emotional, behavioral and physical changes, negatively affecting the mother, child and family life. However, fluoxetine is excreted in breast milk, causing short and long-term effects on children who were exposed to the drug during lactation, so studies that seek to uncover the consequences of these effects are needed. Thus, the aim of this study was to evaluate the effects of fluoxetine on the nutritional characteristics of milk and on growth and neurobehavioral development of the offspring on a rat model. Lactating rats were divided into 4 groups: control group and three experimental groups, which were treated with different doses of fluoxetine (1, 10 and 20 mg/kg) during the lactation. Dams body weight and milk properties were measured, as well as offspring's growth and physical and neurobehavioral development. Results showed that the use of fluoxetine during lactation decreased dam's body weight and alters milk's properties, leading to a decrease in offspring's growth until adulthood. Therefore, the use of fluoxetine during lactation needs to be cautiously evaluated, with the benefits to the mothers and the associated risk to the offspring carefully balance.
Collapse
Affiliation(s)
- Gabriel R Abreu
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Avenida Professor Doutor Orlando Marques de Paiva, 87, 05508-270 São Paulo, Brazil.
| | - Julia Zaccarelli-Magalhães
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Avenida Professor Doutor Orlando Marques de Paiva, 87, 05508-270 São Paulo, Brazil
| | - Laís S Féba
- Health Science Institute, Mackenzie Presbyterian University, Rua da Consolação, 930, 01302-907 São Paulo, Brazil
| | - Emily Y Penna
- Health Science Institute, Mackenzie Presbyterian University, Rua da Consolação, 930, 01302-907 São Paulo, Brazil
| | - Julia P Silva
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Avenida Professor Doutor Orlando Marques de Paiva, 87, 05508-270 São Paulo, Brazil
| | - Mariana V Teixeira
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Avenida Professor Doutor Orlando Marques de Paiva, 87, 05508-270 São Paulo, Brazil
| | - Marianna Manes
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Avenida Professor Doutor Orlando Marques de Paiva, 87, 05508-270 São Paulo, Brazil
| | - Guilherme A Vergara
- Hospital Israelita Albert Einstein, Avenida Albert Einstein, 627, 05652-000 São Paulo, Brazil
| | | | - André R Fukushima
- Centro Universitário das Américas FAM, Rua Augusta, 1508, 01304-001 São Paulo, Brazil; School of Health Sciences IGESP, Rua da Consolação, 1025, 01301-000 São Paulo, Brazil
| | - Esther L Ricci
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Avenida Professor Doutor Orlando Marques de Paiva, 87, 05508-270 São Paulo, Brazil; Health Science Institute, Mackenzie Presbyterian University, Rua da Consolação, 930, 01302-907 São Paulo, Brazil; School of Health Sciences IGESP, Rua da Consolação, 1025, 01301-000 São Paulo, Brazil
| | - Helenice S Spinosa
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Avenida Professor Doutor Orlando Marques de Paiva, 87, 05508-270 São Paulo, Brazil
| |
Collapse
|
17
|
Dong J, Tang Z, Zou L, Zhou Y, Chen J. Visible light-induced hydrogen atom transfer trifluoromethylthiolation of aldehydes with bismuth catalyst. Chem Commun (Camb) 2024; 60:742-745. [PMID: 38116589 DOI: 10.1039/d3cc05048c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
By using a combination of BiCl3 and TBACl as a ligand-to-metal charge transfer (LMCT) photocatalyst, hydrogen atom transfer trifluoromethylthiolation of aldehydes was achieved under visible light irradiation. The present method provides economical and operationally simple access to trifluoromethylthioesters using low toxicity and cost-effective bismuth catalysts under mild reaction conditions. Based on the radical trapping experiments, the direct conversion of aldehydes to acyl radicals via chlorine radicals as HAT reagents was proposed as the reaction mechanism.
Collapse
Affiliation(s)
- Jun Dong
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, Yunnan Minzu University, Yuehua Street, Kunming, 650504, China.
| | - Zhuang Tang
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, Yunnan Minzu University, Yuehua Street, Kunming, 650504, China.
| | - Luqian Zou
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, Yunnan Minzu University, Yuehua Street, Kunming, 650504, China.
| | - Yongyun Zhou
- Yunnan Key Laboratory of Chiral Functional Substance Research and Application, Yunnan Minzu University, Yuehua Street, Kunming, 650504, China.
| | - Jingchao Chen
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, Yunnan Minzu University, Yuehua Street, Kunming, 650504, China.
- Yunnan Key Laboratory of Chiral Functional Substance Research and Application, Yunnan Minzu University, Yuehua Street, Kunming, 650504, China.
| |
Collapse
|
18
|
Dong Y, Lu Z, Gao T, Wei Z, Ou Z, Shi Z, Shen J. A polypeptide derived from pilose antler ameliorates CUMS-induced depression-like behavior by SENP2-PLCβ4 signaling axis. Eur J Pharmacol 2024; 963:176247. [PMID: 38056617 DOI: 10.1016/j.ejphar.2023.176247] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/10/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023]
Abstract
Neurogenesis is known to be closely associated with depression. We aimed to investigate whether a polypeptide monomer derived from pilose antler (polypeptide sequence LSALEGVFYP, PAP) exerts an antidepressant effect by influencing neurogenesis, and to elucidate the mechanism of its antidepressant action. Behavioral tests were performed to observe the antidepressant effect of PAP. Neurogenesis in the dentate gyrus (DG) region of hippocampus was observed by immunofluorescence. The expression of key proteins of Sentrin/SUMO-specific proteases 2 (SENP2)- Phosphoinositide-specific phospholipase C beta 4 (PLCβ4) pathway was accessed by co-immunoprecipitation (Co-IP), and the calcium homeostasis associated proteins were observed via Western blot (WB). Subsequently, temozolomide (TMZ) pharmacologically blocked neurogenesis to verify the antidepressant effect of PAP on neurogenesis. The mechanism of PAP antidepressant effect was verified by constructing a sh-SENP2 virus vector to silence SENP2 protein. Finally, corticosterone (CORT)-induced PC12 cell model was used to verify whether PAP was involved in the process of deconjugated PLCβ4 SUMOylated. The results showed that PAP improved depression-like behavior and neurogenesis induced by chronic unpredictable mild stimulation (CUMS). In addition, PAP acted on SENP2-PLCβ4 pathway to deconjugate the SUMOylation of PLCβ4 and affect calcium homeostasis. Pharmacological blockade of neurogenesis by TMZ treatment impaired the antidepressant efficacy of PAP. Knockout of SENP2 in the CUMS model attenuated the antidepressant response of PAP, and the impaired neurogenesis was not ameliorated by PAP treatment. In summary, PAP acted on the SENP2-PLCβ4 signaling pathway to inhibit the SUMOylation of PLCβ4 and maintain calcium homeostasis, thereby protecting neurogenesis and playing an antidepressant role.
Collapse
Affiliation(s)
- Yu Dong
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Institute of Literature in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zihan Lu
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, 210009, China
| | - Tiantian Gao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhifeng Wei
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhijie Ou
- Neurology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, Jiangsu, 215500, China.
| | - Zheng Shi
- Institute of Literature in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Jie Shen
- Institute of Literature in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
19
|
Orozco-Hernández JM, Elizalde-Velázquez GA, Gómez-Oliván LM, Santamaría-González GO, Rosales-Pérez KE, García-Medina S, Galar-Martínez M, Juan-Reyes NS. Acute exposure to fluoxetine leads to oxidative stress and hematological disorder in Danio rerio adults. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 905:167391. [PMID: 37758136 DOI: 10.1016/j.scitotenv.2023.167391] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/18/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023]
Abstract
Fluoxetine (FLX), a selective serotonin reuptake inhibitor (SSRI), is consistently introduced into the environment due to its ongoing consumption and inadequate removal by wastewater treatment plants. As a result, the scientific community has displayed a keen interest in investigating the potential toxicological effects associated with this medication. Nevertheless, there is a scarcity of available data regarding the impact of FLX on blood parameters. With this in mind, this study aimed to evaluate the potential toxicological consequences of FLX at environmentally significant concentrations (5, 16, and 40 ng/L) following a 96-hour acute exposure blood parameters in Danio rerio fish. Moreover, the investigation encompassed an assessment of oxidative stress parameters to determine whether the drug could induce disruptions in the REDOX status of the fish. The findings unveiled that FLX prompted the induction of oxidative stress in various organs of the fish, encompassing the liver, gut, brain, and gills. Notably, the gills and brain exhibited heightened susceptibility to the drug's effects compared to other organs. Furthermore, following acute exposure to FLX, there was an upregulation of antioxidant-related genes (sod, cat, gpx, nrf1, and nrf2), thereby providing additional evidence supporting the induction of oxidative stress in the organs of the fish. Lastly, FLX significantly impacted the customary values of various blood parameters, including glucose, blood urea nitrogen, alanine aminotransferase, alkaline phosphatase, red blood cell count, hemoglobin, and hematocrit. Thus, it can be inferred that FLX harmed the overall health status of the fish, resulting in the development of liver disease, anemia, and other associated illnesses.
Collapse
Affiliation(s)
- José Manuel Orozco-Hernández
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico
| | - Gustavo Axel Elizalde-Velázquez
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico
| | - Leobardo Manuel Gómez-Oliván
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico.
| | - Guadalupe Ofelia Santamaría-González
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico
| | - Karina Elisa Rosales-Pérez
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico
| | - Sandra García-Medina
- Laboratorio de Toxicología Acuática, Departamento de Farmacia, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Adolfo López Mateos, Av. Wilfrido Massieu s/n y cerrada Manuel Stampa, Col. Industrial Vallejo, Ciudad de México CP, 07700, Mexico
| | - Marcela Galar-Martínez
- Laboratorio de Toxicología Acuática, Departamento de Farmacia, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Adolfo López Mateos, Av. Wilfrido Massieu s/n y cerrada Manuel Stampa, Col. Industrial Vallejo, Ciudad de México CP, 07700, Mexico
| | - Nely San Juan-Reyes
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico
| |
Collapse
|
20
|
Bai Z, Lansbergen B, Ritter T. Bicyclopentylation of Alcohols with Thianthrenium Reagents. J Am Chem Soc 2023; 145:25954-25961. [PMID: 38010346 PMCID: PMC10704608 DOI: 10.1021/jacs.3c10024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 11/29/2023]
Abstract
Herein we present the first method for the synthesis of bicyclo[1.1.1]pentyl (BCP) alkyl ethers from alcohols. The reaction uses BCP-thianthrenium reagents and is catalyzed by a dual copper/photoredox catalyst system. Unlike known alkylations of tertiary alcohols via carbocation intermediates, our Cu-mediated radical process circumvents the labile BCP carbocations. The approach demonstrates a broad tolerance for functional groups when applied to primary, secondary, and even tertiary alcohols. In addition, we highlight the utility of this method in late-stage functionalizations of both natural products and pharmaceuticals as well as in the rapid construction of BCP analogs of known pharmaceuticals that would otherwise be difficult to access.
Collapse
Affiliation(s)
- Zibo Bai
- Max-Planck-Institut für
Kohlenforschung, Kaiser-Wilhelm-Platz 1, D-45470 Mülheim an der Ruhr, Germany
| | - Beatrice Lansbergen
- Max-Planck-Institut für
Kohlenforschung, Kaiser-Wilhelm-Platz 1, D-45470 Mülheim an der Ruhr, Germany
| | - Tobias Ritter
- Max-Planck-Institut für
Kohlenforschung, Kaiser-Wilhelm-Platz 1, D-45470 Mülheim an der Ruhr, Germany
| |
Collapse
|
21
|
Wojtas A. The possible place for psychedelics in pharmacotherapy of mental disorders. Pharmacol Rep 2023; 75:1313-1325. [PMID: 37934320 PMCID: PMC10661751 DOI: 10.1007/s43440-023-00550-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/17/2023] [Accepted: 10/17/2023] [Indexed: 11/08/2023]
Abstract
Since its emergence in the 1960s, the serotonergic theory of depression bore fruit in the discovery of a plethora of antidepressant drugs affecting the lives of millions of patients. While crucial in the history of drug development, recent studies undermine the effectiveness of currently used antidepressant drugs in comparison to placebo, emphasizing the long time it takes to initiate the therapeutic response and numerous adverse effects. Thus, the scope of contemporary pharmacological research shifts from drugs affecting the serotonin system to rapid-acting antidepressant drugs. The prototypical representative of the aforementioned class is ketamine, an NMDA receptor antagonist capable of alleviating the symptoms of depression shortly after the drug administration. This discovery led to a paradigm shift, focusing on amino-acidic neurotransmitters and growth factors. Alas, the drug is not perfect, as its therapeutic effect diminishes circa 2 weeks after administration. Furthermore, it is not devoid of some severe side effects. However, there seems to be another, more efficient, and safer way to target the glutamatergic system. Hallucinogenic agonists of the 5-HT2A receptor, commonly known as psychedelics, are nowadays being reconsidered in clinical practice, shedding their infamous 1970s stigma. More and more clinical studies prove their clinical efficacy and rapid onset after a single administration while bearing fewer side effects. This review focuses on the current state-of-the-art literature and most recent clinical studies concerning the use of psychedelic drugs in the treatment of mental disorders. Specifically, the antidepressant potential of LSD, psilocybin, DMT, and 5-MeO-DMT will be discussed, together with a brief summary of other possible applications.
Collapse
Affiliation(s)
- Adam Wojtas
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland.
| |
Collapse
|
22
|
Tsugiyama LE, Macedo Moraes RC, Cavalcante Moraes YA, Francis-Oliveira J. Promising new pharmacological targets for depression: The search for efficacy. Drug Discov Today 2023; 28:103804. [PMID: 37865307 DOI: 10.1016/j.drudis.2023.103804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/31/2023] [Accepted: 10/16/2023] [Indexed: 10/23/2023]
Abstract
Pharmacological treatment of major depressive disorder (MDD) still relies on the use of serotonergic drugs, despite their limited efficacy. A few mechanistically new drugs have been developed in recent years, but many fail in clinical trials. Several hypotheses have been proposed to explain MDD pathophysiology, indicating that physiological processes such as neuroplasticity, circadian rhythms, and metabolism are potential targets. Here, we review the current state of pharmacological treatments for MDD, as well as the preclinical and clinical evidence for an antidepressant effect of molecules that target non-serotonergic systems. We offer some insights into the challenges facing the development of new antidepressant drugs, and the prospect of finding more effectiveness for each target discussed.
Collapse
Affiliation(s)
- Lucila Emiko Tsugiyama
- Kansai Medical University, Graduate School of Medicine, iPS Cell Applied Medicine, Hirakata, Osaka, Japan
| | - Ruan Carlos Macedo Moraes
- University of Alabama at Birmingham, Department of Psychiatry and Behavioral Neurobiology, Birmingham, AL, USA; Biomedical Sciences Institute, Department of Human Physiology, Sao Paulo University, Sao Paulo, Brazil
| | | | - Jose Francis-Oliveira
- University of Alabama at Birmingham, Department of Psychiatry and Behavioral Neurobiology, Birmingham, AL, USA; Biomedical Sciences Institute, Department of Human Physiology, Sao Paulo University, Sao Paulo, Brazil.
| |
Collapse
|
23
|
Fricke HP, Krajco CJ, Perry MJ, Desorcy‐Scherer KM, Wake LA, Charles JF, Hernandez LL. Developmental fluoxetine exposure affects adolescent and adult bone depending on the dose and period of exposure in mice. Physiol Rep 2023; 11:e15881. [PMID: 38031314 PMCID: PMC10687345 DOI: 10.14814/phy2.15881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 10/11/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023] Open
Abstract
At the end of gestation, fetal skeleton rapidly accumulates calcium, and bone development continues in offspring postnatally. To accommodate, maternal skeletal physiology is modulated in a serotonin-dependent manner. Selective serotonin reuptake inhibitors (SSRIs) are generally considered safe for treatment of major depressive disorder, postpartum depression, and other psychiatric illnesses during the peripartum period, but because serotonin affects bone remodeling, SSRIs are associated with decreased bone mass across all ages and sexes, and the impact of SSRIs during fetal and postnatal development has not been fully investigated. In the present study, our aim was to examine developmental fluoxetine exposure on offspring skeleton and to assess varying degrees of impact depending on dose and window of exposure in short-term and long-term contexts. We established that a low dose of lactational fluoxetine exposure caused a greater degree of insult to offspring bone than either a low dose during fetal and postpartum development or a high dose during lactation only in mice. We further discovered lasting impacts of developmental fluoxetine exposure, especially during lactation only, on adult bone and body composition. Herein, we provide evidence fluoxetine exposure during early development may have detrimental effects on the skeleton of offspring at weaning and into adulthood.
Collapse
Affiliation(s)
- Hannah P. Fricke
- Endocrinology and Reproductive Physiology ProgramUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Department of Animal and Dairy SciencesUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Chandler J. Krajco
- Department of Animal and Dairy SciencesUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Molly J. Perry
- Department of Animal and Dairy SciencesUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Katelyn M. Desorcy‐Scherer
- Department of Animal and Dairy SciencesUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- School of NursingUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Lella A. Wake
- Departments of Orthopedics and MedicineBrigham and Women's HospitalBostonMassachusettsUSA
| | - Julia F. Charles
- Departments of Orthopedics and MedicineBrigham and Women's HospitalBostonMassachusettsUSA
| | - Laura L. Hernandez
- Endocrinology and Reproductive Physiology ProgramUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Department of Animal and Dairy SciencesUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| |
Collapse
|
24
|
Payet JM, Stevens L, Russo AM, Jaehne EJ, van den Buuse M, Kent S, Lowry CA, Baratta MV, Hale MW. The Role of Dorsal Raphe Nucleus Serotonergic Systems in Emotional Learning and Memory in Male BALB/c Mice. Neuroscience 2023; 534:1-15. [PMID: 37852412 DOI: 10.1016/j.neuroscience.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/20/2023]
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are the first-line pharmacological treatment for a variety of anxiety-, trauma- and stressor-related disorders. Although they are efficacious, therapeutic improvements require several weeks of treatment and are often associated with an initial exacerbation of symptoms. The dorsal raphe nucleus (DR) has been proposed as an important target for the modulation of emotional responses and the therapeutic effects of SSRIs. Using a fear-conditioning paradigm we aimed to understand how SSRIs affect emotional learning and memory, and their effects on serotonergic circuitry. Adult male BALB/c mice were treated with vehicle (n = 16) or the SSRI fluoxetine (18 mg/kg/d) acutely (n = 16), or chronically (21d, n = 16), prior to fear conditioning. Treatment was stopped, and half of the mice (n = 8/treatment group) were exposed to cued fear memory recall 72 h later. Activation of DR serotonergic neurons during fear conditioning (Experiment 1) or fear memory recall (Experiment 2), was measured using dual-label immunohistochemistry for Tph2 and c-Fos. Acute and chronic fluoxetine treatment reduced associative fear learning without affecting memory recall and had opposite effects on anxiety-like behaviour. Acute fluoxetine decreased serotonergic activity in the DR, while chronic treatment led to serotonergic activity that was indistinguishable from that of control levels in DRD and DRV subpopulations. Chronic fluoxetine facilitated fear extinction, which was associated with rostral DRD inhibition. These findings provide further evidence that SSRIs can alter aspects of learning and memory processes and are consistent with a role for discrete populations of DR serotonergic neurons in regulating fear- and anxiety-related behaviours.
Collapse
Affiliation(s)
- Jennyfer M Payet
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Laura Stevens
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Adrian M Russo
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Emily J Jaehne
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia; Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Maarten van den Buuse
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Stephen Kent
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Christopher A Lowry
- Department of Integrative Physiology and Centre for Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Michael V Baratta
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Matthew W Hale
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia.
| |
Collapse
|
25
|
Fricke HP, Krajco CJ, Perry MJ, Reisner MA, Brettingen LJ, Wake LA, Charles JF, Hernandez LL. In utero, lactational, or peripartal fluoxetine administration has differential implications on the murine maternal skeleton. Physiol Rep 2023; 11:e15837. [PMID: 37813559 PMCID: PMC10562136 DOI: 10.14814/phy2.15837] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/04/2023] [Accepted: 09/25/2023] [Indexed: 10/12/2023] Open
Abstract
The peripartal period is marked by alterations in calcium metabolism to accommodate for embryonic skeletal mineralization and support bone development of offspring in early life, and serotonin plays a critical role in modulating peripartal bone remodeling. Selective serotonin reuptake inhibitors (SSRIs) are commonly used as first-line treatment for psychiatric illness during pregnancy and the postpartum period and considered safe for maternal use during this time frame. In order to evaluate the effect of peripartal alterations of the serotonergic system on maternal skeletal physiology, we treated dams with the SSRI fluoxetine during gestation only, lactation only, or during the entire peripartal period. Overall, we found a low dose of fluoxetine during gestation only had minimal impacts on maternal bone at weaning, but there were implications on maternal skeleton at weaning when dams were exposed during lactation only or during the entire peripartal period. We found that these effects were differential between female mice dosed lactationally or peripartally, and there were also impacts on maternal mammary gland at weaning in both of these groups. Though SSRIs are largely considered safe maternally during the peripartal period, this study raises the question whether safety of SSRIs, specifically fluoxetine, during the peripartal period should be reevaluated.
Collapse
Affiliation(s)
- Hannah P. Fricke
- Endocrinology and Reproductive Physiology ProgramUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Department of Dairy ScienceUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Chandler J. Krajco
- Department of Dairy ScienceUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Molly J. Perry
- Department of Dairy ScienceUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Maggie A. Reisner
- Department of Dairy ScienceUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | | | - Lella A. Wake
- Departments of Orthopedics and MedicineBrigham and Women's HospitalBostonMassachusettsUSA
| | - Julia F. Charles
- Departments of Orthopedics and MedicineBrigham and Women's HospitalBostonMassachusettsUSA
| | - Laura L. Hernandez
- Endocrinology and Reproductive Physiology ProgramUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Department of Dairy ScienceUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| |
Collapse
|
26
|
Zhan D, Wang X, Zheng Y, Wang S, Yang B, Pan B, Wang N, Wang Z. Integrative dissection of 5-hydroxytryptamine receptors-related signature in the prognosis and immune microenvironment of breast cancer. Front Oncol 2023; 13:1147189. [PMID: 37795441 PMCID: PMC10546427 DOI: 10.3389/fonc.2023.1147189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 08/14/2023] [Indexed: 10/06/2023] Open
Abstract
Background Depression increases the risk of breast cancer recurrence and metastasis. However, there lacks potential biomarkers for predicting prognosis in breast cancer. 5-hydroxytryptamine (5-HT) plays a key role in the pathogenesis and treatment of depression. In this study, we developed a prognostic signature based on 5-HT receptors (5-HTRs) and elucidated its potential immune regulatory mechanisms for breast cancer prognosis. Methods Oncomine, GEPIA, UALCAN, cBioPortal, Kaplan-Meier plotter, and TIMER were used to analyze differential expression, prognostic value, genetic alteration, and immune cell infiltration of HTRs in breast cancer patients. The model training and validation assays were based on the analyses of GSE1456 and GSE86166. A risk signature was established by univariate and multivariate Cox regression analyses. The transwell assay was utilized to verify the effect of the 5-HTRs expression on breast cancer invasion. Effects of HTR2A/2B inhibitor on CD8+ T cell proliferation and infiltration as well as apoptosis of 4T1 cells in the tumor microenvironment were detected by flow cytometry and TUNEL assay. Zebrafish and mouse breast cancer xenografts were used to determine the effect of HTR2A/2B inhibitor on breast cancer metastasis. Results The expression levels of HTR1A, HTR1B, HTR2A, HTR2B, HTR2C, HTR4, and HTR7 were significantly downregulated in highly malignant breast cancer types. 5-HTRs were significantly associated with recurrence-free survival (RFS) in breast cancer patients. The genetic alteration of HTR1D, HTR3A, HTR3B, and HTR6 in breast cancer patients was significantly associated with shorter overall survival (OS). Finally, HTR2A and HTR2B were determined to construct the risk signature. The expression of HTR2A/2B was positively correlated with the infiltration of immune cells such as CD8+ T cells and macrophages. Furthermore, inhibition of HTR2A expression could suppress CD8+ T cell proliferation and enhance invasion and metastasis of breast cancer cells in both zebrafish and mice model. Conclusions The HTR2A/2B risk signature not only highlights the significance of HTRs in breast cancer prognosis by modulating cancer immune microenvironment, but also provides a novel gene-testing tool for early prevention of depression in breast cancer patients and lead to an improved prognosis and quality of life.
Collapse
Affiliation(s)
- Dandan Zhan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuan Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yifeng Zheng
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shengqi Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Bowen Yang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Bo Pan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Neng Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhiyu Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
27
|
Kim J, Kim TE, Lee SH, Koo JW. The Role of Glutamate Underlying Treatment-resistant Depression. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2023; 21:429-446. [PMID: 37424412 PMCID: PMC10335903 DOI: 10.9758/cpn.22.1034] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 07/11/2023]
Abstract
The monoamine hypothesis has significantly improved our understanding of mood disorders and their treatment by linking monoaminergic abnormalities to the pathophysiology of mood disorders. Even 50 years after the monoamine hypothesis was established, some patients do not respond to treatments for depression, including selective serotonin reuptake drugs. Accumulating evidence shows that patients with treatment-resistant depression (TRD) have severe abnormalities in the neuroplasticity and neurotrophic factor pathways, indicating that different treatment approaches may be necessary. Therefore, the glutamate hypothesis is gaining attention as a novel hypothesis that can overcome monoamine restrictions. Glutamate has been linked to structural and maladaptive morphological alterations in several brain areas associated with mood disorders. Recently, ketamine, an N-methyl-D-aspartate receptor (NMDAR) antagonist, has shown efficacy in TRD treatment and has received the U.S. Food and Drug Administration approval, revitalizing psychiatry research. However, the mechanism by which ketamine improves TRD remains unclear. In this review, we re-examined the glutamate hypothesis, bringing the glutamate system onboard to join the modulation of the monoamine systems, emphasizing the most prominent ketamine antidepressant mechanisms, such as NMDAR inhibition and NMDAR disinhibition in GABAergic interneurons. Furthermore, we discuss the animal models used in preclinical studies and the sex differences in the effects of ketamine.
Collapse
Affiliation(s)
- Jeongseop Kim
- Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Korea
| | - Tae-Eun Kim
- Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Korea
| | - Seung-Hwan Lee
- Department of Psychiatry, Inje University Ilsan Paik Hospital, Goyang, Korea
| | - Ja Wook Koo
- Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Korea
| |
Collapse
|
28
|
Frey M, Smigielski L, Tini E, Fekete S, Fleischhaker C, Wewetzer C, Karwautz A, Correll CU, Gerlach M, Taurines R, Plener PL, Malzahn U, Kornbichler S, Weninger L, Brockhaus M, Reuter-Dang SY, Reitzle K, Rock H, Imgart H, Heuschmann P, Unterecker S, Briegel W, Banaschewski T, Fegert JM, Hellenschmidt T, Kaess M, Kölch M, Renner T, Rexroth C, Walitza S, Schulte-Körne G, Romanos M, Egberts KM. Therapeutic Drug Monitoring in Children and Adolescents: Findings on Fluoxetine from the TDM-VIGIL Trial. Pharmaceutics 2023; 15:2202. [PMID: 37765171 PMCID: PMC10534581 DOI: 10.3390/pharmaceutics15092202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/18/2023] [Accepted: 08/19/2023] [Indexed: 09/29/2023] Open
Abstract
Fluoxetine is the recommended first-line antidepressant in many therapeutic guidelines for children and adolescents. However, little is known about the relationships between drug dose and serum level as well as the therapeutic serum reference range in this age group. Within a large naturalistic observational prospective multicenter clinical trial ("TDM-VIGIL"), a transdiagnostic sample of children and adolescents (n = 138; mean age, 15; range, 7-18 years; 24.6% males) was treated with fluoxetine (10-40 mg/day). Analyses of both the last timepoint and all timepoints (n = 292 observations), utilizing (multiple) linear regressions, linear mixed-effect models, and cumulative link (mixed) models, were used to test the associations between dose, serum concentration, outcome, and potential predictors. The receiver operating curve and first to third interquartile methods, respectively, were used to examine concentration cutoff and reference values for responders. A strong positive relationship was found between dose and serum concentration of fluoxetine and its metabolite. Higher body weight was associated with lower serum concentrations, and female sex was associated with lower therapeutic response. The preliminary reference ranges for the active moiety (fluoxetine+norfluoxetine) were 208-328 ng/mL (transdiagnostically) and 201.5-306 ng/mL (depression). Most patients showed marked (45.6%) or minimal (43.5%) improvements and reported no adverse effects (64.9%). This study demonstrated a clear linear dose-serum level relationship for fluoxetine in youth, with the identified reference range being within that established for adults.
Collapse
Affiliation(s)
- Michael Frey
- Faculty of Applied Healthcare Science, Deggendorf Institute of Technology, 94469 Deggendorf, Germany
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, LMU Munich, 80097 Munich, Germany
| | - Lukasz Smigielski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, 8032 Zürich, Switzerland; (L.S.)
| | - Elvira Tini
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, 8032 Zürich, Switzerland; (L.S.)
| | - Stefanie Fekete
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Center for Mental Health, University Hospital of Wuerzburg, 97080 Wuerzburg, Germany
| | - Christian Fleischhaker
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Center Freiburg, 79104 Freiburg, Germany
| | | | - Andreas Karwautz
- Department of Child and Adolescent Psychiatry, Medical University Vienna, 1090 Vienna, Austria
| | - Christoph U. Correll
- Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany
- Department of Psychiatry, The Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY 11004, USA
- Department of Psychiatry and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Manfred Gerlach
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Center for Mental Health, University Hospital of Wuerzburg, 97080 Wuerzburg, Germany
| | - Regina Taurines
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Center for Mental Health, University Hospital of Wuerzburg, 97080 Wuerzburg, Germany
| | - Paul L. Plener
- Department of Child and Adolescent Psychiatry, Medical University Vienna, 1090 Vienna, Austria
- Department of Child and Adolescent Psychiatry/Psychotherapy, University Hospital Ulm, 89075 Ulm, Germany
| | - Uwe Malzahn
- Clinical Trial Center Wuerzburg, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
| | - Selina Kornbichler
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, LMU Munich, 80097 Munich, Germany
| | - Laura Weninger
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, LMU Munich, 80097 Munich, Germany
| | | | - Su-Yin Reuter-Dang
- Specialist Practice and Medical Care Centre for Child and Adolescent Psychiatry Munich, Dr. Epple & Dr. Reuter-Dang, 81241 Munich, Germany
| | - Karl Reitzle
- Specialist Practice and Medical Care Center for Child and Adolescent Psychiatry Munich, 81241 Munich, Germany
| | - Hans Rock
- Central Information Office, Department of Neurology, Philipps University of Marburg, 35112 Marburg, Germany
| | - Hartmut Imgart
- Parkland-Clinic, Clinic for Psychosomatics and Psychotherapy, Academic Teaching Hospital for the University Gießen, 34537 Bad Wildungen, Germany
| | - Peter Heuschmann
- Clinical Trial Center Wuerzburg, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
- Institute of Clinical Epidemiology and Biometry, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Stefan Unterecker
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Wuerzburg, 97080 Wuerzburg, Germany
| | - Wolfgang Briegel
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Center for Mental Health, University Hospital of Wuerzburg, 97080 Wuerzburg, Germany
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Leopoldina Hospital, 97422 Schweinfurt, Germany
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Jörg M. Fegert
- Department of Child and Adolescent Psychiatry/Psychotherapy, University Hospital Ulm, 89075 Ulm, Germany
| | - Tobias Hellenschmidt
- Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatic medicine, Vivantes Clinic Berlin Neukölln, 12351 Berlin, Germany
| | - Michael Kaess
- Clinic for Child and Adolescent Psychiatry, Center for Psychosocial Medicine, University Hospital Heidelberg, 69115 Heidelberg, Germany
- University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, 3000 Bern, Switzerland
| | - Michael Kölch
- Department of Child and Adolescent Psychiatry and Psychotherapy, Brandenburg Medical School Brandenburg, 16816 Neuruppin, Germany
- Department of Child and Adolescent Psychiatry, Neurology, Psychosomatics, and Psychotherapy, University Medical Center Rostock, 18147 Rostock, Germany
| | - Tobias Renner
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Psychiatry and Psychotherapy Tuebingen, Center of Mental Health Tuebingen, 72076 Tuebingen , Germany
| | - Christian Rexroth
- Clinic for Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University of Regensburg at the Regensburg District Hospital, Medbo KU, 93053 Regensburg, Germany
| | - Susanne Walitza
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, 8032 Zürich, Switzerland; (L.S.)
- Zurich Center for Integrative Human Physiology, University of Zurich, 8057 Zürich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH, 8057 Zürich, Switzerland
| | - Gerd Schulte-Körne
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, LMU Munich, 80097 Munich, Germany
| | - Marcel Romanos
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Center for Mental Health, University Hospital of Wuerzburg, 97080 Wuerzburg, Germany
| | - Karin Maria Egberts
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Center for Mental Health, University Hospital of Wuerzburg, 97080 Wuerzburg, Germany
| |
Collapse
|
29
|
Wu Y, Jiang Y, Wang F, Wang B, Chen C. Direct electrophilic and radical isoperfluoropropylation with i-C 3F 7-Iodine(III) reagent (PFPI reagent). Commun Chem 2023; 6:177. [PMID: 37620542 PMCID: PMC10449889 DOI: 10.1038/s42004-023-00986-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023] Open
Abstract
The isoperfluoropropyl group (i-C3F7) is an emerging motif in pharmaceuticals, agrichemicals and functional materials. However, isoperfluoropropylated compounds remain largely underexplored, presumably due to the lack of efficient access to these compounds. Herein, we disclose the practical and efficient isoperfluoropropylation of aromatic C-H bonds through the invention of a hypervalent-iodine-based reagent-PFPI reagent, that proceeds via a Ag-X coupling process. The activation of the PFPI reagent without any catalysts or additives was demonstrated in the synthesis of isoperfluoropropylated electron-rich heterocycles, while its activity under photoredox catalysis was shown in the synthesis of isoperfluoropropylated non-activated arenes. Detailed mechanistic experiments and DFT calculations revealed a SET-induced concerted mechanistic pathway in the photoredox reactions. In addition, the unique conformation of i-C3F7 in products, that involved intramolecular hydrogen bond was investigated by X-ray single-crystal diffraction and variable-temperature NMR experiments.
Collapse
Affiliation(s)
- Yaxing Wu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Yunchen Jiang
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Fei Wang
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Bin Wang
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Chao Chen
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China.
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
30
|
Liu Z, Chen B, Xiang S, Hu S. Self-immolative nanocapsules precisely regulate depressive neuronal microenvironment for synergistic antidepression therapy. J Nanobiotechnology 2023; 21:274. [PMID: 37592281 PMCID: PMC10433581 DOI: 10.1186/s12951-023-02008-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/14/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Pharmacotherapy constitutes the first-line treatment for depression. However, its clinical use is hindered by several limitations, such as time lag, side effects, and narrow therapeutic windows. Nanotechnology can be employed to shorten the onset time by ensuring permeation across the blood brain barrier (BBB) to precisely deliver more therapeutic agents; unfortunately, formidable challenges owing to the intrinsic shortcomings of commercial drugs remain. RESULTS Based on the extraordinary capability of monoamines to regulate the neuronal environment, we engineer a network nanocapsule for delivering serotonin (5-hydroxytryptamine, 5-HT) and catalase (CAT) to the brain parenchyma for synergistic antidepression therapy. The nanoantidepressants are fabricated by the formation of 5-HT polymerization and simultaneous payload CAT, following by surface modifications using human serum albumin and rabies virus glycoprotein. The virus-inspired nanocapsules benefit from the surface-modifying strategies and exhibit pronounced BBB penetration. Once nanocapsules reach the brain parenchyma, the mildly acidic conditions trigger the release of 5-HT from the sacrificial nanocapsule. Releasing 5-HT further positively regulate moods, relieving depressive symptoms. Meanwhile, cargo CAT alleviates neuroinflammation and enhances therapeutic efficacy of 5-HT. CONCLUSION Altogether, the results offer detailed information encouraging the rational designing of nanoantidepressants and highlighting the potential of nanotechnology in mental health disorder therapies.
Collapse
Affiliation(s)
- Ziyao Liu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Biological, Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Bei Chen
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Shijun Xiang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Key Laboratory of Biological, Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
31
|
Joo MK, Kim DH. Vagus nerve-dependent effects of fluoxetine on anxiety- and depression-like behaviors in mice. Eur J Pharmacol 2023:175862. [PMID: 37331682 DOI: 10.1016/j.ejphar.2023.175862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/08/2023] [Accepted: 06/15/2023] [Indexed: 06/20/2023]
Abstract
The vagus nerve is a major pathway in the body that is responsible for regulating the activity of the parasympathetic nervous system, which plays an important role in mood disorders including anxiety and depression. Fluoxetine, also known as Prozac, is widely used to treat depression. Nevertheless, there are few studies on the vagus nerve-mediated action of fluoxetine. In this study, we aimed to investigate the vagus nerve-dependent actions of fluoxetine in mice with restraint stress-induced or antibiotics-induced anxiety- and depression-like behaviors. Compared to sham operation, vagotomy alone did not exhibit significant effects on behavioral changes and serotonin-related biomarkers in mice not exposed to stress, antibiotics, or fluoxetine. Oral administration of fluoxetine significantly alleviated anxiety- and depression-like behaviors. However, celiac vagotomy significantly attenuated the anti-depressive effects of fluoxetine. The vagotomy also inhibited the effect of fluoxetine to attenuate restraint stress- or cefaclor-induced reduction in serotonin levels and Htr1a mRNA expression in the hippocampus. These findings suggest that the vagus nerve may regulate the efficacy of fluoxetine for depression.
Collapse
Affiliation(s)
- Min-Kyung Joo
- Neurobiota Research Center and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul, 02447, South Korea.
| | - Dong-Hyun Kim
- Neurobiota Research Center and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul, 02447, South Korea.
| |
Collapse
|
32
|
Tan H, Martin JM, Alton LA, Lesku JA, Wong BBM. Widespread psychoactive pollutant augments daytime restfulness and disrupts diurnal activity rhythms in fish. CHEMOSPHERE 2023; 326:138446. [PMID: 36940830 DOI: 10.1016/j.chemosphere.2023.138446] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/13/2023] [Accepted: 03/17/2023] [Indexed: 06/18/2023]
Abstract
Pharmaceutical pollution is a major driver of global change, with the capacity to alter key behavioural and physiological traits in exposed animals. Antidepressants are among the most commonly detected pharmaceuticals in the environment. Despite well-documented pharmacological effects of antidepressants on sleep in humans and other vertebrates, very little is known about their ecologically relevant impacts as pollutants on non-target wildlife. Accordingly, we investigated the effects of acute 3-day exposure of eastern mosquitofish (Gambusia holbrooki) to field-realistic levels (nominal concentrations: 30 and 300 ng/L) of the widespread psychoactive pollutant, fluoxetine, on diurnal activity patterns and restfulness, as indicators of disruptions to sleep. We show that exposure to fluoxetine disrupted diel activity patterns, which was driven by augmentation of daytime inactivity. Specifically, unexposed control fish were markedly diurnal, swimming farther during the day and exhibiting longer periods and more bouts of inactivity at night. However, in fluoxetine-exposed fish, this natural diel rhythm was eroded, with no differences in activity or restfulness observed between the day and night. As a misalignment in the circadian rhythm has been shown to adversely affect fecundity and lifespan in animals, our findings reveal a potentially serious threat to the survival and reproductive success of pollutant-exposed wildlife.
Collapse
Affiliation(s)
- Hung Tan
- School of Biological Sciences, Monash University, Melbourne, Australia.
| | - Jake M Martin
- School of Biological Sciences, Monash University, Melbourne, Australia; Department of Wildlife, Fish, and Environmental Studies, Swedish University of Agricultural Sciences, Umeå, Sweden; Department of Zoology, Stockholm University, Stockholm, Sweden
| | - Lesley A Alton
- School of Biological Sciences, Monash University, Melbourne, Australia
| | - John A Lesku
- School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Australia; Research Centre for Future Landscapes, La Trobe University, Melbourne, Australia
| | - Bob B M Wong
- School of Biological Sciences, Monash University, Melbourne, Australia
| |
Collapse
|
33
|
Tseng YT, Zhao B, Ding H, Liang L, Schaefke B, Wang L. Systematic evaluation of a predator stress model of depression in mice using a hierarchical 3D-motion learning framework. Transl Psychiatry 2023; 13:178. [PMID: 37231005 DOI: 10.1038/s41398-023-02481-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/07/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023] Open
Abstract
Investigation of the neurobiology of depression in humans depends on animal models that attempt to mimic specific features of the human disorder. However, frequently-used paradigms based on social stress cannot be easily applied to female mice which has led to a large sex bias in preclinical studies of depression. Furthermore, most studies focus on one or only a few behavioral assessments, with time and practical considerations prohibiting a comprehensive evaluation. In this study, we demonstrate that predator stress effectively induced depression-like behaviors in both male and female mice. By comparing predator stress and social defeat models, we observed that the former elicited a higher level of behavioral despair and the latter elicited more robust social avoidance. Furthermore, the use of machine learning (ML)-based spontaneous behavioral classification can distinguish mice subjected to one type of stress from another, and from non-stressed mice. We show that related patterns of spontaneous behaviors correspond to depression status as measured by canonical depression-like behaviors, which illustrates that depression-like symptoms can be predicted by ML-classified behavior patterns. Overall, our study confirms that the predator stress induced phenotype in mice is a good reflection of several important aspects of depression in humans and illustrates that ML-supported analysis can simultaneously evaluate multiple behavioral alterations in different animal models of depression, providing a more unbiased and holistic approach for the study of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yu-Ting Tseng
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Binghao Zhao
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Hui Ding
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Lisha Liang
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Bernhard Schaefke
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Liping Wang
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
34
|
Fricke HP, Hernandez LL. The Serotonergic System and Bone Metabolism During Pregnancy and Lactation and the Implications of SSRI Use on the Maternal-Offspring Dyad. J Mammary Gland Biol Neoplasia 2023; 28:7. [PMID: 37086330 PMCID: PMC10122632 DOI: 10.1007/s10911-023-09535-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/06/2023] [Indexed: 04/23/2023] Open
Abstract
Lactation is a physiological adaptation of the class Mammalia and is a product of over 200 million years of evolution. During lactation, the mammary gland orchestrates bone metabolism via serotonin signaling in order to provide sufficient calcium for the offspring in milk. The role of serotonin in bone remodeling was first discovered over two decades ago, and the interplay between serotonin, lactation, and bone metabolism has been explored in the years following. It is estimated that postpartum depression affects 10-15% of the population, and selective serotonin reuptake inhibitors (SSRI) are often used as the first-line treatment. Studies conducted in humans, nonhuman primates, sheep, and rodents have provided evidence that there are consequences on both parent and offspring when serotonin signaling is disrupted during the peripartal period; however, the long-term consequences of disruption of serotonin signaling via SSRIs during the peripartal period on the maternal and offspring skeleton are not fully known. This review will focus on the relationship between the mammary gland, serotonin, and bone remodeling during the peripartal period and the skeletal consequences of the dysregulation of the serotonergic system in both human and animal studies.
Collapse
Affiliation(s)
- Hannah P Fricke
- Animal and Dairy Sciences Department, University of Wisconsin-Madison, Madison, WI, USA
| | - Laura L Hernandez
- Animal and Dairy Sciences Department, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
35
|
Talati MN, Vemireddy S, Seelam SD, Halmuthur. M. SK. Synthesis and immunomodulatory activity of novel amino acid analogues of fluoxetine. SYNTHETIC COMMUN 2023. [DOI: 10.1080/00397911.2023.2196024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Affiliation(s)
- Mamta N. Talati
- OSPC Division, Vaccine Immunology Laboratory, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Sravanthi Vemireddy
- OSPC Division, Vaccine Immunology Laboratory, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Siva Deepthi Seelam
- OSPC Division, Vaccine Immunology Laboratory, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Sampath Kumar Halmuthur. M.
- OSPC Division, Vaccine Immunology Laboratory, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
36
|
Kodali M, Jankay T, Shetty AK, Reddy DS. Pathophysiological basis and promise of experimental therapies for Gulf War Illness, a chronic neuropsychiatric syndrome in veterans. Psychopharmacology (Berl) 2023; 240:673-697. [PMID: 36790443 DOI: 10.1007/s00213-023-06319-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/17/2023] [Indexed: 02/16/2023]
Abstract
This article describes the pathophysiology and potential treatments for Gulf War Illness (GWI), which is a chronic neuropsychiatric illness linked to a combination of chemical exposures experienced by service personnel during the first Gulf War in 1991. However, there is currently no effective treatment for veterans with GWI. The article focuses on the current status and efficacy of existing therapeutic interventions in preclinical models of GWI, as well as potential perspectives of promising therapies. GWI stems from changes in brain and peripheral systems in veterans, leading to neurocognitive deficits, as well as physiological and psychological effects resulting from multifaceted changes such as neuroinflammation, oxidative stress, and neuronal damage. Aging not only renders veterans more susceptible to GWI symptoms, but also attenuates their immune capabilities and response to therapies. A variety of experimental models are being used to investigate the pathophysiology and develop therapies that have the ability to alleviate devastating symptoms. Over two dozen therapeutic interventions targeting neuroinflammation, mitochondrial dysfunction, neuronal injury, and neurogenesis are being tested, including agents such as curcumin, curcumin nanoparticles, monosodium luminol, melatonin, resveratrol, fluoxetine, rolipram, oleoylethanolamide, ketamine, levetiracetam, nicotinamide riboside, minocycline, pyridazine derivatives, and neurosteroids. Preclinical outcomes show that some agents have promise, including curcumin, resveratrol, and ketamine, which are being tested in clinical trials in GWI veterans. Neuroprotectants and other compounds such as monosodium luminol, melatonin, levetiracetam, oleoylethanolamide, and nicotinamide riboside appear promising for future clinical trials. Neurosteroids have been shown to have neuroprotective and disease-modifying properties, which makes them a promising medicine for GWI. Therefore, accelerated clinical studies are urgently needed to evaluate and launch an effective therapy for veterans displaying GWI.
Collapse
Affiliation(s)
- Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University School of Medicine, College Station, TX, USA
| | - Tanvi Jankay
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University School of Medicine, College Station, TX, USA.,Texas A&M Health Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, 8447 Riverside Pkwy, Bryan, TX, 77807, USA
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX, USA. .,Texas A&M Health Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, 8447 Riverside Pkwy, Bryan, TX, 77807, USA.
| |
Collapse
|
37
|
Nichols AL, Blumenfeld Z, Luebbert L, Knox HJ, Muthusamy AK, Marvin JS, Kim CH, Grant SN, Walton DP, Cohen BN, Hammar R, Looger L, Artursson P, Dougherty DA, Lester HA. Selective Serotonin Reuptake Inhibitors within Cells: Temporal Resolution in Cytoplasm, Endoplasmic Reticulum, and Membrane. J Neurosci 2023; 43:2222-2241. [PMID: 36868853 PMCID: PMC10072302 DOI: 10.1523/jneurosci.1519-22.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/02/2022] [Accepted: 11/27/2022] [Indexed: 03/05/2023] Open
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are the most prescribed treatment for individuals experiencing major depressive disorder. The therapeutic mechanisms that take place before, during, or after SSRIs bind the serotonin transporter (SERT) are poorly understood, partially because no studies exist on the cellular and subcellular pharmacokinetic properties of SSRIs in living cells. We studied escitalopram and fluoxetine using new intensity-based, drug-sensing fluorescent reporters targeted to the plasma membrane, cytoplasm, or endoplasmic reticulum (ER) of cultured neurons and mammalian cell lines. We also used chemical detection of drug within cells and phospholipid membranes. The drugs attain equilibrium in neuronal cytoplasm and ER at approximately the same concentration as the externally applied solution, with time constants of a few s (escitalopram) or 200-300 s (fluoxetine). Simultaneously, the drugs accumulate within lipid membranes by ≥18-fold (escitalopram) or 180-fold (fluoxetine), and possibly by much larger factors. Both drugs leave cytoplasm, lumen, and membranes just as quickly during washout. We synthesized membrane-impermeant quaternary amine derivatives of the two SSRIs. The quaternary derivatives are substantially excluded from membrane, cytoplasm, and ER for >2.4 h. They inhibit SERT transport-associated currents sixfold or 11-fold less potently than the SSRIs (escitalopram or fluoxetine derivative, respectively), providing useful probes for distinguishing compartmentalized SSRI effects. Although our measurements are orders of magnitude faster than the therapeutic lag of SSRIs, these data suggest that SSRI-SERT interactions within organelles or membranes may play roles during either the therapeutic effects or the antidepressant discontinuation syndrome.SIGNIFICANCE STATEMENT Selective serotonin reuptake inhibitors stabilize mood in several disorders. In general, these drugs bind to SERT, which clears serotonin from CNS and peripheral tissues. SERT ligands are effective and relatively safe; primary care practitioners often prescribe them. However, they have several side effects and require 2-6 weeks of continuous administration until they act effectively. How they work remains perplexing, contrasting with earlier assumptions that the therapeutic mechanism involves SERT inhibition followed by increased extracellular serotonin levels. This study establishes that two SERT ligands, fluoxetine and escitalopram, enter neurons within minutes, while simultaneously accumulating in many membranes. Such knowledge will motivate future research, hopefully revealing where and how SERT ligands engage their therapeutic target(s).
Collapse
Affiliation(s)
- Aaron L Nichols
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91106
| | - Zack Blumenfeld
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91106
- Keck School of Medicine, University of Southern California, Los Angeles, California 90007
| | - Laura Luebbert
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91106
- Institute of Biology, Leiden University, 2333 BE Leiden, The Netherlands
| | - Hailey J Knox
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91106
| | - Anand K Muthusamy
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91106
| | - Jonathan S Marvin
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Viginia 20147
| | - Charlene H Kim
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91106
| | - Stephen N Grant
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91106
| | - David P Walton
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91106
| | - Bruce N Cohen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91106
| | - Rebekkah Hammar
- Department of Pharmacy, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Loren Looger
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Viginia 20147
| | - Per Artursson
- Department of Pharmacy, Uppsala University, SE-751 23 Uppsala, Sweden
- Science for Life Laboratory Drug Discovery and Development Platform and Uppsala University Drug Optimization and Pharmaceutical Profiling Platform, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Dennis A Dougherty
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91106
| | - Henry A Lester
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91106
| |
Collapse
|
38
|
La-Ongthong K, Chantarojsiri T, Soorukram D, Leowanawat P, Reutrakul V, Kuhakarn C. Electrochemical trifluoromethylation of 2-isocyanobiaryls using CF 3SO 2Na: synthesis of 6-(trifluoromethyl)phenanthridines. Org Biomol Chem 2023; 21:4225-4236. [PMID: 36880879 DOI: 10.1039/d3ob00239j] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
An efficient trifluoromethylation of 2-isocyanobiaryls was developed through the constant current electrolysis, employing sodium trifluoromethanesulfinate (CF3SO2Na) as the trifluoromethyl source. The method enabled the syntheses of a series of 6-(trifluoromethyl)phenanthridine derivatives in moderate to high yields under metal- and oxidant-free conditions. A gram-scale synthesis highlights the synthetic versatility of the reported protocol.
Collapse
Affiliation(s)
- Kannika La-Ongthong
- Department of Chemistry, Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand. .,Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand
| | - Teera Chantarojsiri
- Department of Chemistry, Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand. .,Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand
| | - Darunee Soorukram
- Department of Chemistry, Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand. .,Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand
| | - Pawaret Leowanawat
- Department of Chemistry, Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand. .,Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand
| | - Vichai Reutrakul
- Department of Chemistry, Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand. .,Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand
| | - Chutima Kuhakarn
- Department of Chemistry, Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand. .,Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand
| |
Collapse
|
39
|
Lv S, Yao K, Zhang Y, Zhu S. NMDA receptors as therapeutic targets for depression treatment: Evidence from clinical to basic research. Neuropharmacology 2023; 225:109378. [PMID: 36539011 DOI: 10.1016/j.neuropharm.2022.109378] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/08/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
Ketamine, functioning as a channel blocker of the excitatory glutamate-gated N-methyl-d-aspartate (NMDA) receptors, displays compelling fast-acting and sustained antidepressant effects for treatment-resistant depression. Over the past decades, clinical and preclinical studies have implied that the pathology of depression is associated with dysfunction of glutamatergic transmission. In particular, the discovery of antidepressant agents modulating NMDA receptor function has prompted breakthroughs for depression treatment compared with conventional antidepressants targeting the monoaminergic system. In this review, we first summarized the signalling pathway of the ketamine-mediated antidepressant effects, based on the glutamate hypothesis of depression. Second, we reviewed the hypotheses of the synaptic mechanism and network of ketamine antidepressant effects within different brain areas and distinct subcellular localizations, including NMDA receptor antagonism on GABAergic interneurons, extrasynaptic and synaptic NMDA receptor-mediated antagonism, and ketamine blocking bursting activities in the lateral habenula. Third, we reviewed the different roles of NMDA receptor subunits in ketamine-mediated cognitive and psychiatric behaviours in genetically-manipulated rodent models. Finally, we summarized the structural basis of NMDA receptor channel blockers and discussed NMDA receptor modulators that have been reported to exert potential antidepressant effects in animal models or in clinical trials. Integrating the cutting-edge technologies of cryo-EM and artificial intelligence-based drug design (AIDD), we expect that the next generation of first-in-class rapid antidepressants targeting NMDA receptors would be an emerging direction for depression therapeutics. This article is part of the Special Issue on 'Ketamine and its Metabolites'.
Collapse
Affiliation(s)
- Shiyun Lv
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China; University of Chinese Academy of Sciences, Beijing, China
| | - Kejie Yao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China; University of Chinese Academy of Sciences, Beijing, China
| | - Youyi Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China; University of Chinese Academy of Sciences, Beijing, China
| | - Shujia Zhu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
40
|
Rivi V, Benatti C, Rigillo G, Blom JMC. Invertebrates as models of learning and memory: investigating neural and molecular mechanisms. J Exp Biol 2023; 226:jeb244844. [PMID: 36719249 DOI: 10.1242/jeb.244844] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In this Commentary, we shed light on the use of invertebrates as model organisms for understanding the causal and conserved mechanisms of learning and memory. We provide a condensed chronicle of the contribution offered by mollusks to the studies on how and where the nervous system encodes and stores memory and describe the rich cognitive capabilities of some insect species, including attention and concept learning. We also discuss the use of planarians for investigating the dynamics of memory during brain regeneration and highlight the role of stressful stimuli in forming memories. Furthermore, we focus on the increasing evidence that invertebrates display some forms of emotions, which provides new opportunities for unveiling the neural and molecular mechanisms underlying the complex interaction between stress, emotions and cognition. In doing so, we highlight experimental challenges and suggest future directions that we expect the field to take in the coming years, particularly regarding what we, as humans, need to know for preventing and/or delaying memory loss. This article has an associated ECR Spotlight interview with Veronica Rivi.
Collapse
Affiliation(s)
- Veronica Rivi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Cristina Benatti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giovanna Rigillo
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Joan M C Blom
- Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
41
|
Correia D, Domingues I, Faria M, Oliveira M. Effects of fluoxetine on fish: What do we know and where should we focus our efforts in the future? THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 857:159486. [PMID: 36257440 DOI: 10.1016/j.scitotenv.2022.159486] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 06/16/2023]
Abstract
Fluoxetine is one of the most studied and detected selective serotonin reuptake inhibitors in the aquatic environment, found at concentrations ranging from ng/L to μg/L. Its presence in this environment can induce effects on aquatic organisms that may compromise their fitness. Several experimental studies have demonstrated that fluoxetine can induce neurotoxicity, genetic and biochemical changes, and cause behavioral dysfunction in a wide range of fish species. However, contradictory results can be found. There is thus the need for a comprehensive review of the current state of knowledge on the effects of fluoxetine on fish at different levels of biological organization, highlighting inclusive patterns and discussing the potential causes for the contradictory results, that can be found in the available literature. This review also aims to explore and identify the main gaps in knowledge and areas for future research. We conclude that environmentally relevant concentrations of fluoxetine (e.g., from 0.00345 μg/L) produced adverse effects and often this concentration range is not addressed in conventional environmental risk assessment strategies. Its environmental persistence and ionizable properties reinforce the need for standardized testing with representative aquatic models, targeting endpoints sensitive to the specific mode of action of fluoxetine, in order to assess and rank its actual environmental risk to aquatic ecosystems.
Collapse
Affiliation(s)
- Daniela Correia
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - Inês Domingues
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | | | - Miguel Oliveira
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| |
Collapse
|
42
|
Temple NJ. Research strategies in nutrition in health and disease: The failure of mechanistic research. Front Nutr 2023; 10:1082182. [PMID: 36742421 PMCID: PMC9893282 DOI: 10.3389/fnut.2023.1082182] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/02/2023] [Indexed: 01/20/2023] Open
Abstract
This paper critically evaluates different research methods in order to assess their value for establishing which dietary changes are most effective for protecting health and preventing disease. The evidence demonstrates that the combined use of observational studies (mainly cohort studies) and randomized controlled trials (RCTs) is the most successful strategy. Studies of the details of body mechanisms in health and disease (mechanistic research) is another commonly used research strategy. However, much evidence demonstrates that it is a far less successful strategy. In order to support the above conclusions research studies from the following areas are discussed: obesity and dietary fat; heart disease and saturated fat; the Mediterranean diet and cardiovascular disease; type 2 diabetes and dietary fiber; and cancer and micronutrients. While mechanistic research has a poor track record in nutrition, it has achieved some success in other areas of biomedical science. This is shown by examining the role of mechanistic research in the discovery of new drugs.
Collapse
|
43
|
Meade EB, Iwanowicz LR, Neureuther N, LeFevre GH, Kolpin DW, Zhi H, Meppelink SM, Lane RF, Schmoldt A, Mohaimani A, Mueller O, Klaper RD. Transcriptome signatures of wastewater effluent exposure in larval zebrafish vary with seasonal mixture composition in an effluent-dominated stream. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 856:159069. [PMID: 36174698 DOI: 10.1016/j.scitotenv.2022.159069] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 06/16/2023]
Abstract
Wastewater treatment plant (WWTP) effluent-dominated streams provide critical habitat for aquatic and terrestrial organisms but also continually expose them to complex mixtures of pharmaceuticals that can potentially impair growth, behavior, and reproduction. Currently, few biomarkers are available that relate to pharmaceutical-specific mechanisms of action. In the experiment reported in this paper, zebrafish (Danio rerio) embryos at two developmental stages were exposed to water samples from three sampling sites (0.1 km upstream of the outfall, at the effluent outfall, and 0.1 km below the outfall) during base-flow conditions from two months (January and May) of a temperate-region effluent-dominated stream containing a complex mixture of pharmaceuticals and other contaminants of emerging concern. RNA-sequencing identified potential biological impacts and biomarkers of WWTP effluent exposure that extend past traditional markers of endocrine disruption. Transcriptomics revealed changes to a wide range of biological functions and pathways including cardiac, neurological, visual, metabolic, and signaling pathways. These transcriptomic changes varied by developmental stage and displayed sensitivity to variable chemical composition and concentration of effluent, thus indicating a need for stage-specific biomarkers. Some transcripts are known to be associated with genes related to pharmaceuticals that were present in the collected samples. Although traditional biomarkers of endocrine disruption were not enriched in either month, a high estrogenicity signal was detected upstream in May and implicates the presence of unidentified chemical inputs not captured by the targeted chemical analysis. This work reveals associations between bioeffects of exposure, stage of development, and the composition of chemical mixtures in effluent-dominated surface water. The work underscores the importance of measuring effects beyond the endocrine system when assessing the impact of bioactive chemicals in WWTP effluent and identifies a need for non-targeted chemical analysis when bioeffects are not explained by the targeted analysis.
Collapse
Affiliation(s)
- Emma B Meade
- School of Freshwater Sciences, University of Wisconsin-Milwaukee, 600 E. Greenfield Ave, Milwaukee, WI 53204, United States
| | - Luke R Iwanowicz
- U.S. Geological Survey, Eastern Ecological Science Center, 11649 Leetown Road, Kearneysville, WV 25430, United States
| | - Nicklaus Neureuther
- School of Freshwater Sciences, University of Wisconsin-Milwaukee, 600 E. Greenfield Ave, Milwaukee, WI 53204, United States
| | - Gregory H LeFevre
- Department of Civil & Environmental Engineering, University of Iowa, 4105 Seamans Center, Iowa City, IA 52242, United States; IIHR-Hydroscience & Engineering, 100 C. Maxwell Stanley Hydraulics Laboratory, Iowa City, IA 52242, United States
| | - Dana W Kolpin
- U.S. Geological Survey, Central Midwest Water Science Center, 400 S. Clinton St, Rm 269 Federal Building, Iowa City, IA 52240, United States
| | - Hui Zhi
- Department of Civil & Environmental Engineering, University of Iowa, 4105 Seamans Center, Iowa City, IA 52242, United States; IIHR-Hydroscience & Engineering, 100 C. Maxwell Stanley Hydraulics Laboratory, Iowa City, IA 52242, United States
| | - Shannon M Meppelink
- U.S. Geological Survey, Central Midwest Water Science Center, 400 S. Clinton St, Rm 269 Federal Building, Iowa City, IA 52240, United States
| | - Rachael F Lane
- U.S. Geological Survey, Kansas Water Science Center, 1217 Biltmore Dr, Lawrence, KS 66049, United States
| | - Angela Schmoldt
- Great Lakes Genomics Center, University of Wisconsin-Milwaukee, 600 E. Greenfield Ave, Milwaukee, WI 53204, United States
| | - Aurash Mohaimani
- Great Lakes Genomics Center, University of Wisconsin-Milwaukee, 600 E. Greenfield Ave, Milwaukee, WI 53204, United States
| | - Olaf Mueller
- Great Lakes Genomics Center, University of Wisconsin-Milwaukee, 600 E. Greenfield Ave, Milwaukee, WI 53204, United States
| | - Rebecca D Klaper
- School of Freshwater Sciences, University of Wisconsin-Milwaukee, 600 E. Greenfield Ave, Milwaukee, WI 53204, United States; Great Lakes Genomics Center, University of Wisconsin-Milwaukee, 600 E. Greenfield Ave, Milwaukee, WI 53204, United States.
| |
Collapse
|
44
|
Systematic review of studies using platelet serotonin content to assess bioeffect of serotonin reuptake inhibitors at the serotonin transporter. Psychopharmacology (Berl) 2023; 240:1-13. [PMID: 36399187 DOI: 10.1007/s00213-022-06276-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/03/2022] [Indexed: 11/19/2022]
Abstract
RATIONALE Assessment of the bioeffect of serotonin reuptake inhibitors (SRIs, including both selective serotonin reuptake inhibitors (SSRIs) and serotonin-norepinephrine reuptake inhibitors (SNRIs)) at the serotonin transporter (SERT) in patients and healthy controls can have important theoretical and clinical implications. OBJECTIVES Bioeffect at SERT has been assessed by neuroimaging of brain SERT occupancy, through in vitro measurements of platelet serotonin (5-HT) uptake, and by measuring platelet 5-HT content pre- and post-initiation of SRI administration. Studies of platelet 5-HT content were reviewed in order to (1) determine the overall apparent bioeffect of SRIs; (2) compare bioeffect across types of SRIs; (3) compare the three approaches to assessing SRI bioeffect; and (4) determine how the findings might inform clinical practice. METHODS We performed a systematic review of the published studies that measured platelet 5-HT content to assess SRI bioeffect at the platelet SERT. Studies using neuroimaging and in vitro platelet 5-HT uptake to assess SRI bioeffect were reviewed for comparison purposes. RESULTS Clinical doses of SRIs typically resulted in 70-90% reductions in platelet 5-HT content. The observed bioeffect at the platelet SERT appeared similar among different SSRIs and SNRIs. The bioeffect estimations based on platelet 5-HT content were consistent with those obtained using neuroimaging to assess brain SERT occupancy and those based on the in vitro measurement of platelet 5-HT uptake. CONCLUSIONS In general, excellent agreement was seen in the apparent SRI bioeffect (70-90% inhibition) among the platelet 5-HT content studies and across the three bioeffect approaches. Theoretical and practical clinical implications are discussed.
Collapse
|
45
|
Jang DY, Yang B, You MJ, Rim C, Kim HJ, Sung S, Kwon MS. Fluoxetine Decreases Phagocytic Function via REV-ERBα in Microglia. Neurochem Res 2023; 48:196-209. [PMID: 36048349 DOI: 10.1007/s11064-022-03733-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/16/2022] [Accepted: 08/21/2022] [Indexed: 01/11/2023]
Abstract
Although fluoxetine (FLX) is a commonly used drug in psychiatric disorders, such as major depressive disorder, anxiety disorder, panic disorder, and obsessive-compulsive disorder, the mechanism by which FLX exerts its therapeutic effect is not completely understood. In this study, we aimed to determine the possible mechanism by which FLX focuses on microglial phagocytosis. FLX reduced phagocytic function in BV2 cells and increased REV-ERBα without affecting other microglia-related genes, such as inflammation and phagocytosis. Although FLX did not change BMAL1 protein levels, it restricted the nucleocytoplasmic transport (NCT) of BMAL1, leading to its cytosolic accumulation. REV-ERBα antagonist SR8278 rescued the decreased phagocytic activity and restricted NCT of BMAL1. We also found that REV-ERBα mediates the effect of FLX via the inhibition of phospho-ERK (pERK). The ERK inhibitor FR180204 was sufficient to reduce phagocytic function in BV2 cells and restrict the NCT of BMAL1. These results were recapitulated in the primary microglia. In conclusion, we propose that FLX decreases phagocytic function and restricts BMAL1 NCT via REV-ERBα. In addition, ERK inhibition mimics the effects of FLX on microglia.
Collapse
Affiliation(s)
- Da-Yoon Jang
- Department of Pharmacology, School of Medicine, Research Institute for Basic Medical Science, CHA University, CHA BIO COMPLEX, 335 Pangyo, Bundang-Gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea.,Research Competency Milestones Program (RECOMP) of School of Medicine, CHA University, Seongnam-si, South Korea
| | - Bohyun Yang
- Department of Pharmacology, School of Medicine, Research Institute for Basic Medical Science, CHA University, CHA BIO COMPLEX, 335 Pangyo, Bundang-Gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Min-Jung You
- Department of Pharmacology, School of Medicine, Research Institute for Basic Medical Science, CHA University, CHA BIO COMPLEX, 335 Pangyo, Bundang-Gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Chan Rim
- Department of Pharmacology, School of Medicine, Research Institute for Basic Medical Science, CHA University, CHA BIO COMPLEX, 335 Pangyo, Bundang-Gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Hui-Ju Kim
- Department of Pharmacology, School of Medicine, Research Institute for Basic Medical Science, CHA University, CHA BIO COMPLEX, 335 Pangyo, Bundang-Gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Soyoung Sung
- Department of Pharmacology, School of Medicine, Research Institute for Basic Medical Science, CHA University, CHA BIO COMPLEX, 335 Pangyo, Bundang-Gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Min-Soo Kwon
- Department of Pharmacology, School of Medicine, Research Institute for Basic Medical Science, CHA University, CHA BIO COMPLEX, 335 Pangyo, Bundang-Gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
46
|
TCA and SSRI Antidepressants Exert Selection Pressure for Efflux-Dependent Antibiotic Resistance Mechanisms in Escherichia coli. mBio 2022; 13:e0219122. [PMID: 36374097 PMCID: PMC9765716 DOI: 10.1128/mbio.02191-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Microbial diversity is reduced in the gut microbiota of animals and humans treated with selective serotonin reuptake inhibitors (SSRIs) and tricyclic antidepressants (TCAs). The mechanisms driving the changes in microbial composition, while largely unknown, is critical to understand considering that the gut microbiota plays important roles in drug metabolism and brain function. Using Escherichia coli, we show that the SSRI fluoxetine and the TCA amitriptyline exert strong selection pressure for enhanced efflux activity of the AcrAB-TolC pump, a member of the resistance-nodulation-cell division (RND) superfamily of transporters. Sequencing spontaneous fluoxetine- and amitriptyline-resistant mutants revealed mutations in marR and lon, negative regulators of AcrAB-TolC expression. In line with the broad specificity of AcrAB-TolC pumps these mutants conferred resistance to several classes of antibiotics. We show that the converse also occurs, as spontaneous chloramphenicol-resistant mutants displayed cross-resistance to SSRIs and TCAs. Chemical-genomic screens identified deletions in marR and lon, confirming the results observed for the spontaneous resistant mutants. In addition, deletions in 35 genes with no known role in drug resistance were identified that conferred cross-resistance to antibiotics and several displayed enhanced efflux activities. These results indicate that combinations of specific antidepressants and antibiotics may have important effects when both are used simultaneously or successively as they can impose selection for common mechanisms of resistance. Our work suggests that selection for enhanced efflux activities is an important factor to consider in understanding the microbial diversity changes associated with antidepressant treatments. IMPORTANCE Antidepressants are prescribed broadly for psychiatric conditions to alter neuronal levels of synaptic neurotransmitters such as serotonin and norepinephrine. Two categories of antidepressants are selective serotonin reuptake inhibitors (SSRIs) and tricyclic antidepressants (TCAs); both are among the most prescribed drugs in the United States. While it is well-established that antidepressants inhibit reuptake of neurotransmitters there is evidence that they also impact microbial diversity in the gastrointestinal tract. However, the mechanisms and therefore biological and clinical effects remain obscure. We demonstrate antidepressants may influence microbial diversity through strong selection for mutant bacteria with increased AcrAB-TolC activity, an efflux pump that removes antibiotics from cells. Furthermore, we identify a new group of genes that contribute to cross-resistance between antidepressants and antibiotics, several act by regulating efflux activity, underscoring overlapping mechanisms. Overall, this work provides new insights into bacterial responses to antidepressants important for understanding antidepressant treatment effects.
Collapse
|
47
|
Yu X, Yao H, Zhang X, Liu L, Liu S, Dong Y. Comparison of LPS and MS-induced depressive mouse model: behavior, inflammation and biochemical changes. BMC Psychiatry 2022; 22:590. [PMID: 36064335 PMCID: PMC9443001 DOI: 10.1186/s12888-022-04233-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 08/29/2022] [Indexed: 12/28/2022] Open
Abstract
Depression is a mental disease involving complex pathophysiological mechanisms, and there are many ways to establish depressive mouse models. The purpose of this study is to comprehensively compare the behavioral changes and its mechanism induced by two different models. This study established two depressive mouse models by maternal separation (MS) or lipopolysaccharide (LPS) administration, and added fluoxetine treatment group respectively for comparison. MS induced more apparent anxiety-like behavior while LPS induced more apparent depressive-like behavior. LPS increased peripheral inflammatory factors more apparent, which were mitigated by fluoxetine. MS inhibited the 5-HT system more obviously and was relieved by fluoxetine. LPS triggered stronger immune response in the hippocampus and prefrontal cortex (PFC). MS significantly reduced the expression of neurotrophic proteins and was alleviated by fluoxetine. Overall, LPS induced stronger system inflammation, while MS impaired the function of HPA axis and 5-HT system. Our results will contribute to a deeper understanding of the pathophysiology of different stress-induced depression and will also help researchers select appropriate models of depression for their own needs.
Collapse
Affiliation(s)
- Xiaojin Yu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning, 110004, P. R. China.
| | - Hui Yao
- grid.412449.e0000 0000 9678 1884Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110012 P. R. China
| | - Xiaohui Zhang
- grid.412467.20000 0004 1806 3501Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004 P. R. China
| | - Lulu Liu
- grid.412467.20000 0004 1806 3501Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004 P. R. China
| | - Shuangmei Liu
- grid.412467.20000 0004 1806 3501Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004 P. R. China
| | - Youjing Dong
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning, 110004, P. R. China.
| |
Collapse
|
48
|
Arun R, Stiniya S, Saranya PV, Anilkumar G. An Overview of Palladium-catalyzed Trifluoromethylation Reactions. J Organomet Chem 2022. [DOI: 10.1016/j.jorganchem.2022.122492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
49
|
Guo R, Zhang X, Bu X, Wang M, Zhao B, Gao Y, Jia Q, Wang Y. Se
‐(Fluoromethyl) Benzenesulfonoselenoates: Shelf‐Stable, Easily Available Reagents for Monofluoromethylselenolation. Chemistry 2022; 28:e202200981. [DOI: 10.1002/chem.202200981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Rui‐Li Guo
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education School of Foreign Languages College of Chemistry & Materials Science Northwest University Xi'an 710069 P. R. China
| | - Xing‐Long Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education School of Foreign Languages College of Chemistry & Materials Science Northwest University Xi'an 710069 P. R. China
| | - Xian‐Pan Bu
- Ankang R&D Center for Se-enriched Products, Key Laboratory of Se-enriched Products Development and Quality Control Ministry of Agriculture and Rural Affairs Ankang Shaanxi 725000 P. R. China
| | - Meng‐Yue Wang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education School of Foreign Languages College of Chemistry & Materials Science Northwest University Xi'an 710069 P. R. China
| | - Bao‐Yin Zhao
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education School of Foreign Languages College of Chemistry & Materials Science Northwest University Xi'an 710069 P. R. China
| | - Ya‐Ru Gao
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education School of Foreign Languages College of Chemistry & Materials Science Northwest University Xi'an 710069 P. R. China
| | - Qiong Jia
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education School of Foreign Languages College of Chemistry & Materials Science Northwest University Xi'an 710069 P. R. China
| | - Yong‐Qiang Wang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education School of Foreign Languages College of Chemistry & Materials Science Northwest University Xi'an 710069 P. R. China
| |
Collapse
|
50
|
Barreto A, Santos J, Capitão A, Eusébio R, Pinheiro Damasceno É, Luísa Machado A, Rocha LS, Calisto V, Amorim MJB, Maria VL. Assessment of diphenhydramine toxicity - Is its mode of action conserved between human and zebrafish? ENVIRONMENT INTERNATIONAL 2022; 164:107263. [PMID: 35504231 DOI: 10.1016/j.envint.2022.107263] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 06/14/2023]
Abstract
The main aim of the study is to evaluate the effects of the pharmaceutical diphenhydramine (DPH) on embryo-larvae Danio rerio across distinct levels of organization - individual and subcellular - and correlate those effects with the DPH mode of action (MoA) assessed by in silico analysis. An embryos heartbeat rate reduction was observed at 10 mg/L DPH, but 0.001 to 10 mg/L did not significantly affect the zebrafish survival, hatching and morphology. Larvae swimming distance decreased (hypoactivity) at 1 and 10 mg/L DPH. Moreover, the straightforward movements decrease and the increase in the zigzag movements or movements with direction changes, shown an erratic swimming behavior. Energy budgets decreased for lipid (0.01 mg/L DPH) and carbohydrate (10 mg/L DPH) contents. Cholinesterase (neural function) and glutathione S-transferase (Phase II biotransformation/antioxidant processes) increased their activities at 10 mg/L DPH, where a decrease in the total glutathione content (antioxidant system) was observed. DNA damage was found at 0.01 and 10 mg/L DPH. However, a DNA repair occurred after subsequent 72 h in clean media. The in silico study revealed a relevant conservation between human and zebrafish DPH target molecules. These data provide a valuable ecotoxicological information about the DPH effects and MoA to non-target organisms.
Collapse
Affiliation(s)
- Angela Barreto
- Department of Biology & CESAM, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Joana Santos
- Department of Biology & CESAM, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ana Capitão
- Department of Biology & CESAM, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Rodrigo Eusébio
- Department of Biology & CESAM, University of Aveiro, 3810-193 Aveiro, Portugal
| | | | - Ana Luísa Machado
- Department of Biology & CESAM, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Luciana S Rocha
- Department of Chemistry & CESAM, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Vânia Calisto
- Department of Chemistry & CESAM, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Mónica J B Amorim
- Department of Biology & CESAM, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Vera L Maria
- Department of Biology & CESAM, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|