Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.789
Filter
Add more filters

Publication year range
1.
Mar Drugs ; 22(7)2024 Jun 30.
Article in English | MEDLINE | ID: mdl-39057418

ABSTRACT

The current 2019-2021 marine pharmacology literature review provides a continuation of previous reviews covering the period 1998 to 2018. Preclinical marine pharmacology research during 2019-2021 was published by researchers in 42 countries and contributed novel mechanism-of-action pharmacology for 171 structurally characterized marine compounds. The peer-reviewed marine natural product pharmacology literature reported antibacterial, antifungal, antiprotozoal, antituberculosis, and antiviral mechanism-of-action studies for 49 compounds, 87 compounds with antidiabetic and anti-inflammatory activities that also affected the immune and nervous system, while another group of 51 compounds demonstrated novel miscellaneous mechanisms of action, which upon further investigation, may contribute to several pharmacological classes. Thus, in 2019-2021, a very active preclinical marine natural product pharmacology pipeline provided novel mechanisms of action as well as new lead chemistry for the clinical marine pharmaceutical pipeline targeting the therapy of several disease categories.


Subject(s)
Anti-Inflammatory Agents , Antitubercular Agents , Antiviral Agents , Aquatic Organisms , Biological Products , Hypoglycemic Agents , Humans , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/chemistry , Biological Products/pharmacology , Biological Products/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antitubercular Agents/pharmacology , Antitubercular Agents/chemistry , Nervous System/drug effects , Immune System/drug effects , Antifungal Agents/pharmacology , Antifungal Agents/chemistry , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry
2.
Annu Rev Physiol ; 82: 177-202, 2020 02 10.
Article in English | MEDLINE | ID: mdl-31738670

ABSTRACT

Endocrine disrupting chemicals are common in our environment and act on hormone systems and signaling pathways to alter physiological homeostasis. Gestational exposure can disrupt developmental programs, permanently altering tissues with impacts lasting into adulthood. The brain is a critical target for developmental endocrine disruption, resulting in altered neuroendocrine control of hormonal signaling, altered neurotransmitter control of nervous system function, and fundamental changes in behaviors such as learning, memory, and social interactions. Human cohort studies reveal correlations between maternal/fetal exposure to endocrine disruptors and incidence of neurodevelopmental disorders. Here, we summarize the major literature findings of endocrine disruption of neurodevelopment and concomitant changes in behavior by four major endocrine disruptor classes:bisphenol A, polychlorinated biphenyls, organophosphates, and polybrominated diphenyl ethers. We specifically review studies of gestational and/or lactational exposure to understand the effects of early life exposure to these compounds and summarize animal studies that help explain human correlative data.


Subject(s)
Behavior/drug effects , Endocrine Disruptors/adverse effects , Nervous System/growth & development , Prenatal Exposure Delayed Effects/pathology , Adult , Animals , Behavior, Animal/drug effects , Benzhydryl Compounds/adverse effects , Female , Humans , Nervous System/drug effects , Phenols/adverse effects , Polybrominated Biphenyls/adverse effects , Polychlorinated Biphenyls/adverse effects , Pregnancy
3.
Int J Mol Sci ; 25(12)2024 Jun 09.
Article in English | MEDLINE | ID: mdl-38928091

ABSTRACT

Pain management in neonates continues to be a challenge. Diverse therapies are available that cause loss of pain sensitivity. However, because of side effects, the search for better options remains open. Dexmedetomidine is a promising drug; it has shown high efficacy with a good safety profile in sedation and analgesia in the immature nervous system. Though dexmedetomidine is already in use for pain control in neonates (including premature neonates) and infants as an adjunct to other anesthetics, the question remains whether it affects the neuronal activity patterning that is critical for development of the immature nervous system. In this study, using the neonatal rat as a model, the pharmacodynamic effects of dexmedetomidine on the nervous and cardiorespiratory systems were studied. Our results showed that dexmedetomidine has pronounced analgesic effects in the neonatal rat pups, and also weakly modified both the immature network patterns of cortical and hippocampal activity and the physiology of sleep cycles. Though the respiration and heart rates were slightly reduced after dexmedetomidine administration, it might be considered as the preferential independent short-term therapy for pain management in the immature and developing brain.


Subject(s)
Animals, Newborn , Dexmedetomidine , Dexmedetomidine/pharmacology , Animals , Rats , Analgesics, Non-Narcotic/pharmacology , Analgesia/methods , Pain Management/methods , Male , Rats, Sprague-Dawley , Pain/drug therapy , Heart Rate/drug effects , Female , Nervous System/drug effects , Nervous System/growth & development
4.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38892402

ABSTRACT

In day-to-day living, individuals are exposed to various environmentally hazardous substances that have been associated with diverse diseases. Exposure to air pollutants can occur during breathing, posing a considerable risk to those with environmental health vulnerabilities. Among vulnerable individuals, maternal exposure can negatively impact the mother and child in utero. The developing fetus is particularly vulnerable to environmentally hazardous substances, with potentially greater implications. Among air pollutants, toluene is neurotoxic, and its effects have been widely explored. However, the impact of low-level toluene exposure in daily life remains unclear. Herein, we evaluated 194 mothers and infants from the Growing children's health and Evaluation of Environment (GREEN) cohort to determine the possible effects of early-life toluene exposure on the nervous system. Using Omics experiments, the effects of toluene were confirmed based on epigenetic changes and altered mRNA expression. Various epigenetic changes were identified, with upregulated expression potentially contributing to diseases such as glioblastoma and Alzheimer's, and downregulated expression being associated with structural neuronal abnormalities. These findings were detected in both maternal and infant groups, suggesting that maternal exposure to environmental hazardous substances can negatively impact the fetus. Our findings will facilitate the establishment of environmental health policies, including the management of environmentally hazardous substances for vulnerable groups.


Subject(s)
Maternal Exposure , Toluene , Humans , Toluene/toxicity , Female , Infant , Maternal Exposure/adverse effects , Pregnancy , Adult , Nervous System/drug effects , Nervous System/embryology , Nervous System/metabolism , Nervous System/growth & development , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Epigenesis, Genetic/drug effects , Male , Mothers , Air Pollutants/toxicity , Infant, Newborn
5.
Zhonghua Yu Fang Yi Xue Za Zhi ; 58(6): 917-923, 2024 Jun 06.
Article in Zh | MEDLINE | ID: mdl-38955742

ABSTRACT

Persistent Organic Pollutants (POPs) have the characteristics of resistance to environmental degradation, bioaccumulation and long-distance migration potential. Maternal exposure to POPs during pregnancy can enter the fetal blood circulation through the placental barrier, and have a potential impact on the functional development of the nervous system of the offspring. This in turn leads to the occurrence and development of neurological defects and diseases in adulthood. The purpose of this paper is to elucidate the effects of exposure to three major POPs (organochlorine compounds, perfluoroalkyl and polyfluoroalkyl substances, and polybrominated diphenyl ethers) during pregnancy on the functional development of the nervous system (social emotions, cognition, language, exercise, and adaptability) in children, and to provide reference for subsequent studies.


Subject(s)
Nervous System , Persistent Organic Pollutants , Prenatal Exposure Delayed Effects , Pregnancy , Humans , Female , Child , Nervous System/drug effects , Nervous System/growth & development , Maternal Exposure/adverse effects , Halogenated Diphenyl Ethers/toxicity , Hydrocarbons, Chlorinated , Child Development/drug effects , Environmental Pollutants/toxicity
6.
Environ Res ; 221: 115295, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36681143

ABSTRACT

BACKGROUND: The combined effects of multiple environmental toxicants and social stressor exposures are widely recognized as important public health problems, likely contributing to health inequities. However, US policy makers at state and federal levels typically focus on one stressor exposure at a time and have failed to develop comprehensive strategies to reduce multiple co-occurring exposures, mitigate cumulative risks and prevent harm. This research aimed to move from considering disparate environmental stressors in isolation to mapping the links between environmental, economic, social and health outcomes as a dynamic complex system using children's exposure to neurodevelopmental toxicants as an illustrative example. Such a model can be used to support a broad range of child developmental and environmental health policy stakeholders in improving their understanding of cumulative effects of multiple chemical, physical, biological and social environmental stressors as a complex system through a collaborative learning process. METHODS: We used system dynamics (SD) group model building to develop a qualitative causal theory linking multiple interacting streams of social stressors and environmental neurotoxicants impacting children's neurodevelopment. A 2 1/2-day interactive system dynamics workshop involving experts across multiple disciplines was convened to develop the model followed by qualitative survey on system insights. RESULTS: The SD causal map covered seven interconnected themes: environmental exposures, social environment, health status, education, employment, housing and advocacy. Potential high leverage intervention points for reducing disparities in children's cumulative neurotoxicant exposures and effects were identified. Workshop participants developed deeper level of understanding about the complexity of cumulative environmental health risks, increased their agreement about underlying causes, and enhanced their capabilities for integrating diverse forms of knowledge about the complex multi-level problem of cumulative chemical and non-chemical exposures. CONCLUSION: Group model building using SD can lead to important insights to into the sociological, policy, and institutional mechanisms through which disparities in cumulative impacts are transmitted, resisted, and understood.


Subject(s)
Environmental Exposure , Models, Biological , Nervous System , Neurotoxins , Child , Humans , Environmental Health , Health Status , Housing , Social Environment , Neurotoxins/toxicity , Nervous System/drug effects , Nervous System/growth & development
7.
J Neuroinflammation ; 19(1): 289, 2022 Dec 03.
Article in English | MEDLINE | ID: mdl-36463233

ABSTRACT

BACKGROUND: Neuroinflammation is one of the most important processes in secondary injury after traumatic brain injury (TBI). Triggering receptor expressed on myeloid cells 2 (TREM2) has been proven to exert neuroprotective effects in neurodegenerative diseases and stroke by modulating neuroinflammation, and promoting phagocytosis and cell survival. However, the role of TREM2 in TBI has not yet been elucidated. In this study, we are the first to use COG1410, an agonist of TREM2, to assess the effects of TREM2 activation in a murine TBI model. METHODS: Adult male wild-type (WT) C57BL/6 mice and adult male TREM2 KO mice were subjected to different treatments. TBI was established by the controlled cortical impact (CCI) method. COG1410 was delivered 1 h after CCI via tail vein injection. Western blot analysis, immunofluorescence, laser speckle contrast imaging (LSCI), neurological behaviour tests, brain electrophysiological monitoring, Evans blue assays, magnetic resonance imaging (MRI), and brain water content measurement were performed in this study. RESULTS: The expression of endogenous TREM2 peaked at 3 d after CCI, and it was mainly expressed on microglia and neurons. We found that COG1410 improved neurological functions within 3 d, as well as neurological functions and brain electrophysiological activity at 2 weeks after CCI. COG1410 exerted neuroprotective effects by inhibiting neutrophil infiltration and microglial activation, and suppressing neuroinflammation after CCI. In addition, COG1410 treatment alleviated blood brain barrier (BBB) disruption and brain oedema; furthermore, COG1410 promoted cerebral blood flow (CBF) recovery at traumatic injury sites after CCI. In addition, COG1410 suppressed neural apoptosis at 3 d after CCI. TREM2 activation upregulated p-Akt, p-CREB, BDNF, and Bcl-2 and suppressed TNF-α, IL-1ß, Bax, and cleaved caspase-3 at 3 d after CCI. Moreover, TREM2 knockout abolished the effects of COG1410 on vascular phenotypes and microglial states. Finally, the neuroprotective effects of COG1410 were suppressed by TREM2 depletion. CONCLUSIONS: Altogether, we are the first to demonstrate that TREM2 activation by COG1410 alleviated neural damage through activation of Akt/CREB/BDNF signalling axis in microglia after CCI. Finally, COG1410 treatment improved neurological behaviour and brain electrophysiological activity after CCI.


Subject(s)
Brain Injuries, Traumatic , Animals , Male , Mice , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/immunology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/immunology , Membrane Glycoproteins/agonists , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Receptors, Immunologic/agonists , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Disease Models, Animal , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/genetics , Neuroinflammatory Diseases/immunology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/immunology , Nervous System/drug effects , Nervous System/immunology
8.
Reprod Biol Endocrinol ; 20(1): 19, 2022 Jan 26.
Article in English | MEDLINE | ID: mdl-35081973

ABSTRACT

BACKGROUND: Nitric oxide and GnRH are biological factors that participate in the regulation of reproductive functions. To our knowledge, there are no studies that link NO and GnRH in the sympathetic ganglia. Thus, the aim of the present work was to investigate the influence of NO on GnRH release from the coeliac ganglion and its effect on luteal regression at the end of pregnancy in the rat. METHODS: The ex vivo system composed by the coeliac ganglion, the superior ovarian nerve, and the ovary of rats on day 21 of pregnancy was incubated for 180 min with the addition, into the ganglionic compartment, of L-NG-nitro arginine methyl ester (L-NAME), a non-selective NO synthase inhibitor. The control group consisted in untreated organ systems. RESULTS: The addition of L-NAME in the coeliac ganglion compartment decreased NO as well as GnRH release from the coeliac ganglion. In the ovarian compartment, and with respect to the control group, we observed a reduced release of GnRH, NO, and noradrenaline, but an increased production of progesterone, estradiol, and expression of their limiting biosynthetic enzymes, 3ß-HSD and P450 aromatase, respectively. The inhibition of NO production by L-NAME in the coeliac ganglion compartment also reduced luteal apoptosis, lipid peroxidation, and nitrotyrosine, whereas it increased the total antioxidant capacity within the corpora lutea. CONCLUSION: Collectively, the results indicate that NO production by the coeliac ganglion modulates the physiology of the ovary and luteal regression during late pregnancy in rats.


Subject(s)
Corpus Luteum/innervation , Corpus Luteum/metabolism , Gonadotropin-Releasing Hormone/metabolism , Nitric Oxide/metabolism , Animals , Drug Interactions , Female , Ganglia, Sympathetic/drug effects , Ganglia, Sympathetic/metabolism , Gestational Age , Gonadotropin-Releasing Hormone/pharmacology , Nervous System/drug effects , Nervous System/metabolism , Neural Pathways/drug effects , Neural Pathways/metabolism , Nitric Oxide/pharmacology , Ovary/innervation , Ovary/metabolism , Pregnancy , Rats
9.
Ecotoxicol Environ Saf ; 242: 113896, 2022 Sep 01.
Article in English | MEDLINE | ID: mdl-35870347

ABSTRACT

Nanoparticles are widely used in industry and personal care, and they inevitably end up in people's bodies and the environment. The widespread use of nanoparticles has raised new concerns about their neurotoxicity, as nanoparticles can enter the nervous system by blood-brain barrier. In neurotoxicity testing, the zebrafish provides powerful tools to overcome the limitations of other models. This paper will provide a comprehensive review of the power of zebrafish in neurotoxicity tests and the neurotoxic effects of nanoparticles, including inorganic, organic, and metal-based nanoparticles, on zebrafish from different perspectives. Such information can be used to predict not only the effects of nanoparticles on other species exposed to the aquatic environment but also the neurotoxicity of nanoparticles in humans.


Subject(s)
Nanoparticles , Nervous System , Animals , Blood-Brain Barrier , Humans , Nanoparticles/toxicity , Nervous System/drug effects , Zebrafish
10.
Int J Mol Sci ; 23(3)2022 Feb 07.
Article in English | MEDLINE | ID: mdl-35163796

ABSTRACT

Developmental neurotoxicity (DNT) of chemical compounds disrupts the formation of a normal brain. There is impressive progress in the development of alternative testing methods for DNT potential in chemicals, some of which also incorporate invertebrate animals. This review briefly touches upon studies on the genetically tractable model organisms of Caenorhabditis elegans and Drosophila melanogaster about the action of specific developmental neurotoxicants. The formation of a functional nervous system requires precisely timed axonal pathfinding to the correct cellular targets. To address this complex key event, our lab developed an alternative assay using a serum-free culture of intact locust embryos. The first neural pathways in the leg of embryonic locusts are established by a pair of afferent pioneer neurons which use guidance cues from membrane-bound and diffusible semaphorin proteins. In a systematic approach according to recommendations for alternative testing, the embryo assay quantifies defects in pioneer navigation after exposure to a panel of recognized test compounds for DNT. The outcome indicates a high predictability for test-compound classification. Since the pyramidal neurons of the mammalian cortex also use a semaphorin gradient for neurite guidance, the assay is based on evolutionary conserved cellular mechanisms, supporting its relevance for cortical development.


Subject(s)
Nervous System/growth & development , Neurotoxicity Syndromes/etiology , Animals , Axon Guidance/drug effects , Disease Models, Animal , Invertebrates , Nervous System/drug effects , Toxicity Tests
11.
J Neurosci ; 40(18): 3631-3645, 2020 04 29.
Article in English | MEDLINE | ID: mdl-32245827

ABSTRACT

N-methyl-D-aspartate receptors (NMDARs) are glutamate-gated ion channels that play critical roles in neuronal development and nervous system function. Here, we developed a model to study NMDARs in early development in zebrafish, by generating CRISPR-mediated lesions in the NMDAR genes, grin1a and grin1b, which encode the obligatory GluN1 subunits. While receptors containing grin1a or grin1b show high Ca2+ permeability, like their mammalian counterpart, grin1a is expressed earlier and more broadly in development than grin1b Both grin1a-/- and grin1b-/- zebrafish are viable. Unlike in rodents, where the grin1 knockout is embryonic lethal, grin1 double-mutant fish (grin1a-/-; grin1b-/-), which lack all NMDAR-mediated synaptic transmission, survive until ∼10 d dpf (days post fertilization), providing a unique opportunity to explore NMDAR function during development and in generating behaviors. Many behavioral defects in the grin1 double-mutant larvae, including abnormal evoked responses to light and acoustic stimuli, prey-capture deficits, and a failure to habituate to acoustic stimuli, are replicated by short-term treatment with the NMDAR antagonist MK-801, suggesting that they arise from acute effects of compromised NMDAR-mediated transmission. Other defects, however, such as periods of hyperactivity and alterations in place preference, are not phenocopied by MK-801, suggesting a developmental origin. Together, we have developed a unique model to study NMDARs in the developing vertebrate nervous system.SIGNIFICANCE STATEMENT Rapid communication between cells in the nervous system depends on ion channels that are directly activated by chemical neurotransmitters. One such ligand-gated ion channel, the NMDAR, impacts nearly all forms of nervous system function. It has been challenging, however, to study the prolonged absence of NMDARs in vertebrates, and hence their role in nervous system development, due to experimental limitations. Here, we demonstrate that zebrafish lacking all NMDAR transmission are viable through early development and are capable of a wide range of stereotypic behaviors. As such, this zebrafish model provides a unique opportunity to study the role of NMDAR in the development of the early vertebrate nervous system.


Subject(s)
Nervous System/embryology , Nervous System/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Zebrafish Proteins/metabolism , Acoustic Stimulation/methods , Animals , Animals, Genetically Modified , Excitatory Amino Acid Antagonists/pharmacology , Female , HEK293 Cells , Humans , Male , Nervous System/drug effects , Photic Stimulation/methods , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Zebrafish , Zebrafish Proteins/antagonists & inhibitors
12.
J Biol Chem ; 295(18): 6120-6137, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32229587

ABSTRACT

The developing nervous system is remarkably sensitive to environmental signals, including disruptive toxins, such as polybrominated diphenyl ethers (PBDEs). PBDEs are an environmentally pervasive class of brominated flame retardants whose neurodevelopmental toxicity mechanisms remain largely unclear. Using dissociated cortical neurons from embryonic Rattus norvegicus, we found here that chronic exposure to 6-OH-BDE-47, one of the most prevalent hydroxylated PBDE metabolites, suppresses both spontaneous and evoked neuronal electrical activity. On the basis of our previous work on mitogen-activated protein kinase (MAPK)/extracellular signal-related kinase (ERK) (MEK) biology and our observation that 6-OH-BDE-47 is structurally similar to kinase inhibitors, we hypothesized that certain hydroxylated PBDEs mediate neurotoxicity, at least in part, by impairing the MEK-ERK axis of MAPK signal transduction. We tested this hypothesis on three experimental platforms: 1) in silico, where modeling ligand-protein docking suggested that 6-OH-BDE-47 is a promiscuous ATP-competitive kinase inhibitor; 2) in vitro in dissociated neurons, where 6-OH-BDE-47 and another specific hydroxylated BDE metabolite similarly impaired phosphorylation of MEK/ERK1/2 and activity-induced transcription of a neuronal immediate early gene; and 3) in vivo in Drosophila melanogaster, where developmental exposures to 6-OH-BDE-47 and a MAPK inhibitor resulted in offspring displaying similarly increased frequency of mushroom-body ß-lobe midline crossing, a metric of axonal guidance. Taken together, our results support that certain ortho-hydroxylated PBDE metabolites are promiscuous kinase inhibitors and can cause disruptions of critical neurodevelopmental processes, including neuronal electrical activity, pre-synaptic functions, MEK-ERK signaling, and axonal guidance.


Subject(s)
Ethers/chemistry , Ethers/pharmacology , Halogenation , Nervous System/growth & development , Neurons/cytology , Neurons/drug effects , Signal Transduction/drug effects , Animals , Drosophila melanogaster , Hydroxylation , Intracellular Space/drug effects , Intracellular Space/metabolism , Nervous System/cytology , Nervous System/drug effects , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology
13.
Pflugers Arch ; 473(12): 1851-1858, 2021 12.
Article in English | MEDLINE | ID: mdl-34633524

ABSTRACT

Global warming and connected acidification of the world ocean attract a substantial amount of research efforts, in particular in a context of their impact on behaviour and metabolism of marine organisms, such as Cnidaria. Nevertheless, mechanisms underlying Cnidarians' neural signalling and behaviour and their (possible) alterations due to the world ocean acidification remain poorly understood. Here we researched for the first time modulation of GABAA receptors (GABAARs) in Actinia equina (Cnidaria: Anthozoa) by pH fluctuations within a range predicted by the world ocean acidification scenarios for the next 80-100 years and by selective pharmacological activation. We found that in line with earlier studies on vertebrates, both changes of pH and activation of GABAARs with a selective allosteric agonist (diazepam) modulate electrical charge transfer through GABAAR and the whole-cell excitability. On top of that, diazepam modifies the animal behavioural reaction on startle response. However, despite behavioural reactions displayed by living animals are controlled by GABAARs, changes of pH do not alter them significantly. Possible mechanisms underlying the species resistance to acidification impact are discussed.


Subject(s)
Aquatic Organisms/metabolism , Nervous System/metabolism , Receptors, GABA-A/metabolism , Sea Anemones/metabolism , Animals , Aquatic Organisms/drug effects , Behavior, Animal/drug effects , Diazepam/pharmacology , Global Warming , Hydrogen-Ion Concentration , Nervous System/drug effects , Sea Anemones/drug effects
14.
J Neurochem ; 157(3): 508-519, 2021 05.
Article in English | MEDLINE | ID: mdl-33421129

ABSTRACT

Brain metabolism evolves rapidly during early post-natal development in the rat. While changes in amino acids, energy metabolites, antioxidants or metabolites involved in phospholipid metabolism have been reported in the early stages, neurometabolic changes during the later post-natal period are less well characterized. Therefore, we aimed to assess the neurometabolic changes in male Wistar rats between post-natal days 29 and 77 (p29-p77) using longitudinal magnetic resonance spectroscopy (MRS) in vivo at 9.4 Tesla. 1 H MRS was performed in the hippocampus between p29 and p77 at 1-week intervals (n = 7) and in the cerebellum between p35 and p77 at 2-week intervals (n = 7) using the SPECIAL sequence at ultra-short echo-time. NOE enhanced and 1 H decoupled 31 P MR spectra were acquired at p35, p48 and p63 (n = 7) in a larger voxel covering cortex, hippocampus and part of the striatum. The hippocampus showed a decrease in taurine concentration and an increase in glutamate (with more pronounced changes until p49), seemingly a continuation of their well-described changes in the early post-natal period. A constant increase in myo-inositol and choline-containing compounds in the hippocampus (in particular glycero-phosphocholine as shown by 31 P MRS) was measured throughout the observation period, probably related to membrane metabolism and myelination. The cerebellum showed only a significant increase in myo-inositol between p35 and p77. In conclusion, this study showed important changes in brain metabolites in both the hippocampus and cerebellum in the later post-natal period (p29/p35-p77) of male rats, something previously unreported. Based on these novel data, changes in some neurometabolites beyond p28-35, conventionally accepted as the cut off for adulthood, should be taken into account in both experimental design and data interpretation in this animal model.


Subject(s)
Nervous System/growth & development , Nervous System/metabolism , Anesthesia/adverse effects , Anesthetics, Inhalation/adverse effects , Animals , Cerebellum/drug effects , Cerebellum/growth & development , Cerebellum/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Choline/metabolism , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/growth & development , Hippocampus/metabolism , Inositol/metabolism , Isoflurane/adverse effects , Magnetic Resonance Spectroscopy , Male , Nervous System/drug effects , Phosphorus Isotopes , Protons , Rats , Rats, Wistar , Taurine/metabolism
15.
Toxicol Appl Pharmacol ; 417: 115477, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33667508

ABSTRACT

N-Ethylpentylone (NEP) is one of the most recent novel stimulants, and there is limited understanding of its toxicity. Here we employed zebrafish model for analyzing the effects of NEP on early embryos and cardiovascular and nervous systems at late developmental stages. We first observed multi-malformations in early embryos and larvae after NEP administration, together with significant deregulations of brain and heart development-associated genes (neurog1, her6, elavl3, nkx2.5, nppa, nppb, tnnt2a) at transcriptional level. Low-dosed NEP treatment induced an anxiety-like phenotype in zebrafish larvae, while higher doses of NEP exerted an inhibitory effect on locomotion and heart rate. Besides, the expression of th (tyrosine hydroxylase) and th2 (tyrosine hydroxylase 2), identifying dopamine (DA) release, were significantly increased during one-hour free swimming after effective low-dosed NEP administration, along with the upregulation of gene fosab and fosb related to stress and anxiety response. D1R antagonist SCH23390 and D2R antagonist sulpiride partially alleviated the aberrances of locomotion and heart rate, indicating dopaminergic receptors were involved in the bidirectional dosage-dependent pattern of NEP-induced performance. Meanwhile, sulpiride offset the upregulated expression of th, th2 and fosab in the group of 1.5 µM NEP, which highlighted the significant role of D2R in NEP-induced locomotive effects. This study systematically described the developmental, neuronal and cardiac toxicity of NEP in zebrafish, and identified the dopaminergic receptors as one of the downstream effectors of NEP administration.


Subject(s)
Benzodioxoles/toxicity , Butylamines/toxicity , Cardiovascular System/drug effects , Dopamine Agonists/toxicity , Dopamine/metabolism , Nervous System/drug effects , Receptors, Dopamine D1/agonists , Receptors, Dopamine D2/agonists , Zebrafish Proteins/agonists , Animals , Animals, Genetically Modified , Cardiovascular System/embryology , Cardiovascular System/metabolism , Female , Gene Expression Regulation, Developmental , Heart Rate/drug effects , Larva/drug effects , Larva/metabolism , Locomotion/drug effects , Male , Nervous System/embryology , Nervous System/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Transcription, Genetic , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
16.
Chem Res Toxicol ; 34(9): 1948-1952, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34491731

ABSTRACT

Exposure to polychlorinated biphenyls (PCBs) is implicated in adverse neurotoxic outcomes. However, the impact of PCBs on the adolescent nervous system has received inadequate attention. We conducted a comprehensive review to identify studies of neurotoxic outcomes following PCB exposure during the adolescent period in rodents. Only four papers were found to meet all inclusion criteria. PCB exposure in adolescent rats caused disruptions in the main functions of the prefrontal cortex, resulting in cognitive deficits. This comprehensive review demonstrates that more research is needed to characterize how PCB exposure adversely affects the adolescent nervous system.


Subject(s)
Nervous System/drug effects , Polychlorinated Biphenyls/toxicity , Adolescent , Animals , Humans
17.
Crit Rev Toxicol ; 51(3): 209-216, 2021 03.
Article in English | MEDLINE | ID: mdl-33905310

ABSTRACT

Mefloquine, a potent blood schizontocide, is effective against drug-resistant Plasmodium falciparum. This property, along with its unique pharmacokinetic profile, makes mefloquine a widely prescribed antimalarial drug. However, several epidemiological studies have raised concerns on the safety of mefloquine as prophylaxis for malaria. Well-documented side-effects of mefloquine include abnormal dreams, insomnia, anxiety, and depressed mood, as well as nausea and dizziness (the last two most frequent effects). The mechanisms that underlie the neurological/psychiatric complications of mefloquine are poorly understood. The aim of this study was to review the literature on the neurotoxic mechanisms of action of mefloquine to better understand its potential toxicity in the central nervous system, highlighting the mechanisms that lead to its psychiatric disorders. Experimental studies on the neurotoxic effects of mefloquine discussed herein include brain transporters of mefloquine, alteration in neurotransmitters, disruption on calcium (Ca2+) homeostasis and neuroinflammation, generation of oxidative stress response in neurons (involving glutathione, increased F2-isoprostanes, accumulation of cytosolic lipid globules), and alteration of voltage-dependent channels, as well as gap junction intercellular communications. Although several hypotheses have been proposed for the mechanisms that mediate mefloquine-induced brain damage, they are not fully understood, necessitating additional studies in the future.


Subject(s)
Antimalarials/toxicity , Mefloquine/toxicity , Nervous System/drug effects , Central Nervous System , Humans
18.
Crit Rev Toxicol ; 51(4): 328-358, 2021 04.
Article in English | MEDLINE | ID: mdl-34074207

ABSTRACT

The current understanding of thyroid-related adverse outcome pathways (AOPs) with adverse neurodevelopmental outcomes in mammals has been reviewed. This served to establish if standard rodent toxicity test methods and in vitro assays allow identifying thyroid-related modes-of-action potentially leading to adverse neurodevelopmental outcomes, and the human relevance of effects - in line with the European Commission's Endocrine Disruptor Criteria. The underlying hypothesis is that an understanding of the key events of relevant AOPs provides insight into differences in incidence, magnitude, or species sensitivity of adverse outcomes. The rodent studies include measurements of serum thyroid hormones, thyroid gland pathology and neurodevelopmental assessments, but do not directly inform on specific modes-of-action. Opportunities to address additional non-routine parameters reflecting critical events of AOPs in toxicological assessments are presented. These parameters appear relevant to support the identification of specific thyroid-related modes-of-action, provided that prevailing technical limitations are overcome. Current understanding of quantitative key event relationships is often weak, but would be needed to determine if the triggering of a molecular initiating event will ultimately result in an adverse outcome. Also, significant species differences in all processes related to thyroid hormone signalling are evident, but the biological implications thereof (including human relevance) are often unknown. In conclusion, careful consideration of the measurement (e.g. timing, method) and interpretation of additional non-routine parameters is warranted. These findings will be used in a subsequent paper to propose a testing strategy to identify if a substance may elicit maternal thyroid hormone imbalance and potentially also neurodevelopmental effects in the progeny.


Subject(s)
Toxicity Tests/methods , Adverse Outcome Pathways , Animals , Endocrine Disruptors , Humans , Nervous System/drug effects , Nervous System/growth & development , Neurotoxicity Syndromes , Risk Assessment , Thyroid Gland , Thyroid Hormones
19.
Pharmacol Res ; 170: 105700, 2021 08.
Article in English | MEDLINE | ID: mdl-34087351

ABSTRACT

Cytisine, a natural bioactive compound that is mainly isolated from plants of the Leguminosae family (especially the seeds of Laburnum anagyroides), has been marketed in central and eastern Europe as an aid in the clinical management of smoking cessation for more than 50 years. Its main targets are neuronal nicotinic acetylcholine receptors (nAChRs), and pre-clinical studies have shown that its interactions with various nAChR subtypes located in different areas of the central and peripheral nervous systems are neuroprotective, have a wide range of biological effects on nicotine and alcohol addiction, regulate mood, food intake and motor activity, and influence the autonomic and cardiovascular systems. Its relatively rigid conformation makes it an attractive template for research of new derivatives. Recent studies of structurally modified cytisine have led to the development of new compounds and for some of them the biological activities are mediated by still unidentified targets other than nAChRs, whose mechanisms of action are still being investigated. The aim of this review is to describe and discuss: 1) the most recent pre-clinical results obtained with cytisine in the fields of neurological and non-neurological diseases; 2) the effects and possible mechanisms of action of the most recent cytisine derivatives; and 3) the main areas warranting further research.


Subject(s)
Alkaloids/pharmacology , Nervous System/drug effects , Receptors, Nicotinic/drug effects , Smoking Cessation Agents/pharmacology , Smoking Cessation , Alkaloids/pharmacokinetics , Alkaloids/toxicity , Animals , Azocines/pharmacokinetics , Azocines/pharmacology , Azocines/toxicity , Humans , Molecular Structure , Nervous System/metabolism , Quinolizines/pharmacokinetics , Quinolizines/pharmacology , Quinolizines/toxicity , Receptors, Nicotinic/metabolism , Smoking Cessation Agents/pharmacokinetics , Smoking Cessation Agents/toxicity , Structure-Activity Relationship
20.
Bioorg Med Chem ; 39: 116131, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33852975

ABSTRACT

Cannabidiol (CBD) and dihydroartemisinin (DHA) can alleviate neuroinflammatory responses. However, they show cytotoxicity, which severely limits their therapeutic windows. Therefore, there is a great need to develop neuroprotective agents with improved safety. Drug-drug conjugate is an emerging approach for enhancing therapeutic index. Herein, the development, synthesis, and the pharmacological characterization of CBD-DHA conjugates were performed. Meanwhile, the combination of CBD and DHA as separate entities was also quantitatively analyzed for direct comparison with CBD-DHA conjugates. In this study, BV-2 microglial cell line was used to mimic primary microglia and the effects of CBD, DHA, the combination of CBD and DHA, as well as CBD-DHA conjugates on LPS-activated signaling molecules and pro-inflammatory factors were assessed. The interaction of CBD and DHA in inhibiting LPS-induced nitric oxide (NO) production was found to be additive. In contrast, DHA was found to synergize with CBD in inhibiting BV-2 cellular viability which implies that the combination of CBD and DHA amplifies their cytotoxicity. CBD-DHA conjugate C3D eliminated the cytotoxicity associated with single CBD/DHA use without significantly compromising the anti-neuroinflammation activity. C3D was more potent than C2D and C4D in inhibiting LPS-induced NO and mRNAs of iNOS and IL-1ß, which implies that the linker length is critical for CBD-DHA conjugates' anti-inflammatory activities. Further signaling characterizations showed that C3D inhibited LPS-induced NF-κB but not MAPKs activation in BV-2 cells, therefore blocking LPS-induced neuroinflammation. This work provides a good example that conjugated drug-drug approach may improve the therapeutic index by increasing the maximum tolerated concentration/dose compared to traditional combination strategy.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Artemisinins/chemistry , Cannabidiol/chemistry , Inflammation/prevention & control , Nervous System/drug effects , Animals , Artemisinins/pharmacology , Cannabidiol/pharmacology , Cell Line , Drug Interactions , Drug Therapy, Combination , Nervous System/pathology
SELECTION OF CITATIONS
SEARCH DETAIL