Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 457
Filter
1.
N Engl J Med ; 390(5): 421-431, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38294974

ABSTRACT

BACKGROUND: Niemann-Pick disease type C is a rare lysosomal storage disorder. We evaluated the safety and efficacy of N-acetyl-l-leucine (NALL), an agent that potentially ameliorates lysosomal and metabolic dysfunction, for the treatment of Niemann-Pick disease type C. METHODS: In this double-blind, placebo-controlled, crossover trial, we randomly assigned patients 4 years of age or older with genetically confirmed Niemann-Pick disease type C in a 1:1 ratio to receive NALL for 12 weeks, followed by placebo for 12 weeks, or to receive placebo for 12 weeks, followed by NALL for 12 weeks. NALL or matching placebo was administered orally two to three times per day, with patients 4 to 12 years of age receiving weight-based doses (2 to 4 g per day) and those 13 years of age or older receiving a dose of 4 g per day. The primary end point was the total score on the Scale for the Assessment and Rating of Ataxia (SARA; range, 0 to 40, with lower scores indicating better neurologic status). Secondary end points included scores on the Clinical Global Impression of Improvement, the Spinocerebellar Ataxia Functional Index, and the Modified Disability Rating Scale. Crossover data from the two 12-week periods in each group were included in the comparisons of NALL with placebo. RESULTS: A total of 60 patients 5 to 67 years of age were enrolled. The mean baseline SARA total scores used in the primary analysis were 15.88 before receipt of the first dose of NALL (60 patients) and 15.68 before receipt of the first dose of placebo (59 patients; 1 patient never received placebo). The mean (±SD) change from baseline in the SARA total score was -1.97±2.43 points after 12 weeks of receiving NALL and -0.60±2.39 points after 12 weeks of receiving placebo (least-squares mean difference, -1.28 points; 95% confidence interval, -1.91 to -0.65; P<0.001). The results for the secondary end points were generally supportive of the findings in the primary analysis, but these were not adjusted for multiple comparisons. The incidence of adverse events was similar with NALL and placebo, and no treatment-related serious adverse events occurred. CONCLUSIONS: Among patients with Niemann-Pick disease type C, treatment with NALL for 12 weeks led to better neurologic status than placebo. A longer period is needed to determine the long-term effects of this agent in patients with Niemann-Pick disease type C. (Funded by IntraBio; ClinicalTrials.gov number, NCT05163288; EudraCT number, 2021-005356-10.).


Subject(s)
Central Nervous System Agents , Niemann-Pick Disease, Type C , Humans , Data Collection , Double-Blind Method , Leucine/analogs & derivatives , Leucine/therapeutic use , Niemann-Pick Disease, Type C/complications , Niemann-Pick Disease, Type C/diagnosis , Niemann-Pick Disease, Type C/drug therapy , Niemann-Pick Disease, Type C/genetics , Treatment Outcome , Cross-Over Studies , Child, Preschool , Child , Adolescent , Young Adult , Adult , Middle Aged , Aged , Central Nervous System Agents/administration & dosage , Central Nervous System Agents/therapeutic use
2.
Psychoneuroendocrinology ; 143: 105823, 2022 09.
Article in English | MEDLINE | ID: mdl-35689985

ABSTRACT

Procedural learning is a vital brain function that allows us to acquire motor skills during development or re-learn them after lesions affecting the motor system. Procedural learning can be improved by feedback of different valence, e.g., monetary or social, mediated by dopaminergic circuits. While processing motivationally relevant stimuli, dopamine interacts closely with oxytocin, whose effects on procedural learning, particularly feedback-based approaches, remain poorly understood. In a randomized, double-blind, placebo-controlled trial, we investigated whether oxytocin modulates the differential effects of monetary and social feedback on procedural learning. Sixty-one healthy male participants were randomized to receive a placebo or oxytocin intranasally. The participants then performed a modified serial reaction time task. Oxytocin plasma concentrations were measured before and after applying the placebo or verum. Groups did not differ regarding general reaction times or measures of procedural learning. For the placebo group, monetary feedback improved procedural learning compared to a neutral control condition. In contrast, the oxytocin group did not show a differential effect of monetary or social feedback despite a significant increase in oxytocin plasma levels after intranasal application. The data suggest that oxytocin does not influence procedural learning per se. Instead, oxytocin seems to attenuate the effects of monetary feedback on procedural learning specifically.


Subject(s)
Central Nervous System Agents , Feedback, Psychological , Learning , Oxytocin , Psychomotor Performance , Reward , Administration, Intranasal , Central Nervous System Agents/administration & dosage , Central Nervous System Agents/pharmacology , Double-Blind Method , Feedback, Psychological/drug effects , Feedback, Psychological/physiology , Humans , Learning/drug effects , Learning/physiology , Male , Oxytocin/administration & dosage , Oxytocin/pharmacology , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Reaction Time , Social Behavior
3.
Neuropsychopharmacology ; 47(11): 1901-1912, 2022 10.
Article in English | MEDLINE | ID: mdl-35396500

ABSTRACT

Atypical responses to sensory stimuli are considered as a core aspect and early life marker of autism spectrum disorders (ASD). Although recent findings performed in mouse ASD genetic models report sensory deficits, these were explored exclusively during juvenile or adult period. Whether sensory dysfunctions might be present at the early life stage and rescued by therapeutic strategy are fairly uninvestigated. Here we found that under cool environment neonatal mice lacking the autism-associated gene Magel2 present pup calls hypo-reactivity and are retrieved with delay by their wild-type dam. This neonatal atypical sensory reactivity to cool stimuli was not associated with autonomic thermoregulatory alteration but with a deficit of the oxytocinergic system. Indeed, we show in control neonates that pharmacogenetic inactivation of hypothalamic oxytocin neurons mimicked atypical thermosensory reactivity found in Magel2 mutants. Furthermore, pharmacological intranasal administration of oxytocin to Magel2 neonates was able to rescue both the atypical thermosensory response and the maternal pup retrieval. This preclinical study establishes for the first-time early life impairments in thermosensory integration and suggest a therapeutic potential benefit of intranasal oxytocin treatment on neonatal atypical sensory reactivity for autism.


Subject(s)
Autistic Disorder , Hypesthesia , Maternal Behavior , Oxytocin , Proteins , Administration, Intranasal , Age Factors , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Autism Spectrum Disorder/complications , Autistic Disorder/complications , Autistic Disorder/genetics , Autistic Disorder/metabolism , Central Nervous System Agents/administration & dosage , Central Nervous System Agents/metabolism , Female , Hypesthesia/etiology , Hypesthesia/genetics , Hypesthesia/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Maternal Behavior/physiology , Mice , Oxytocin/administration & dosage , Oxytocin/metabolism , Proteins/genetics , Proteins/metabolism , Social Behavior
4.
Biol Pharm Bull ; 44(10): 1391-1398, 2021.
Article in English | MEDLINE | ID: mdl-34602547

ABSTRACT

Ultrasound and microbubbles, an ultrasound contrast agent, have recently increased attention to developing novel drug delivery systems. Ultrasound exposure can induce mechanical effects derived from microbubbles behaviors such as an expansion, contraction, and collapse depending on ultrasound conditions. These mechanical effects induce several biological effects, including enhancement of vascular permeability. For drug delivery, one promising approach is enhancing vascular permeability using ultrasound and microbubbles, resulting in improved drug transport to targeted tissues. This approach is applied to several tissues and drugs to cure diseases. This review describes the enhancement of vascular permeability by ultrasound and microbubbles and its therapeutic application, including our recent study. We also discuss the current situation of the field and its potential future perspectives.


Subject(s)
Antineoplastic Agents/administration & dosage , Central Nervous System Agents/administration & dosage , Contrast Media/pharmacology , Drug Delivery Systems/methods , Microbubbles , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/radiation effects , Capillary Permeability/drug effects , Capillary Permeability/radiation effects , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/radiation effects , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/pathology , Humans , Neoplasms/blood supply , Neoplasms/drug therapy , Neoplasms/pathology , Ultrasonography , Xenograft Model Antitumor Assays
5.
Medicine (Baltimore) ; 100(43): e27664, 2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34713861

ABSTRACT

ABSTRACT: This study examined changes in psychotropic medication use associated with the early months of the coronavirus disease 2019 (COVID-19) pandemic. Using Kaiser Permanente Northern California electronic health records, the authors identified adult patients with fills for psychotropic medications and a non-psychotropic comparator (statins) in the 13 weeks before and after the first-known COVID-19-related death in California (March 4, 2020). Generalized estimating equations were used to derive relative risk ratios (RRR) for medication fills compared with the prior year. Analyses were stratified by new and continued fills and patient characteristics. Among 2,405,824 patients, the mean (SD) age was 49.8 (17.9) years; 52.9% were female; 47.9% identified as White; 8.0% and 7.9% had anxiety and depression disorder diagnoses, respectively. Accounting for secular trends, in the 13 weeks following March 4, 2020, there were increased fills for trazodone (RRR = 1.03, 95% CI = 1.02, 1.04), decreased fills for benzodiazepines (RRR = 0.95, 95% CI = 0.94, 0.96) and hypnotics (RRR = 0.97, 95% CI = 0.96, 0.99), and stable fills for antidepressants (RRR = 1.00, 95% CI = 0.99, 1.00). Relative rates of new fills decreased across most medication classes and continued fills either remained stable or demonstrated non-clinically significant decreases. Patients aged ≥65 years demonstrated decreased fills for most medication classes. In the first 13 weeks of the COVID-19 pandemic, fills for most psychotropic medications remained constant or showed small changes relative to the previous year. Continued (compared with new) fills accounted for observed increases in some medication classes. Older adults demonstrated decreased fills of most medications.


Subject(s)
COVID-19/epidemiology , Central Nervous System Agents/administration & dosage , Drug Prescriptions/statistics & numerical data , Adult , Age Factors , Aged , California/epidemiology , Female , Humans , Male , Middle Aged , Pandemics , SARS-CoV-2 , Sex Factors , Socioeconomic Factors
6.
Biomed Pharmacother ; 143: 112117, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34479020

ABSTRACT

Drug delivery to central nervous system (CNS) diseases is very challenging since the presence of the innate blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier that impede drug delivery. Among new strategies to overcome these limitations and successfully deliver drugs to the CNS, nanotechnology-based drug delivery platform, offers potential therapeutic approach for the treatment of some common neurological disorders like Alzheimer's disease, frontotemporal dementia, amyotrophic lateral sclerosis, Parkinson's disease, Huntington's disease. This review aimed to highlight advances in research on the development of nano-based therapeutics for their implications in therapy of CNS disorders. The challenges during clinical translation of nanomedicine from bench to bed side is also discussed.


Subject(s)
Central Nervous System Agents/administration & dosage , Central Nervous System Diseases/drug therapy , Drug Carriers , Nanomedicine , Nanoparticles , Animals , Blood-Brain Barrier/metabolism , Central Nervous System Agents/chemistry , Central Nervous System Agents/metabolism , Central Nervous System Diseases/metabolism , Drug Compounding , Drug Development , Drug Discovery , Humans , Permeability , Translational Research, Biomedical
7.
Behav Brain Res ; 413: 113438, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34224762

ABSTRACT

The rodent caudate-putamen is a large heterogeneous neural structure with distinct anatomical connections that differ in their control of learning processes. Previous research suggests that the anterior and posterior dorsomedial caudate-putamen (a- and p-dmCPu) differentially regulate associative learning with a non-contingent nicotine stimulus. The current study used bilateral NMDA-induced excitotoxic lesions to the a-dmCPu and p-dmCPu to determine the functional involvement of a-dmCPu and p-dmCPu in appetitive learning with contingent nicotine stimulus. Rats with a-dmCPu, p-dmCPu, or sham lesions were trained to lever-press for intravenous nicotine (0.03 mg/kg/inf) followed by access to sucrose 30 s later. After 1, 3, 9, and 20 nicotine-sucrose training sessions, appetitive learning in the form of a goal-tracking response was assessed using a non-contingent nicotine-alone test. All rats acquired nicotine self-administration and learned to retrieve sucrose from a receptacle at equal rates. However, rats with lesions to p-dmCPu demonstrated blunted learning of the nicotine-sucrose association. Our primary findings show that rats with lesions to p-dmCPu had a blunted goal-tracking response to a non-contingent nicotine administration after 20 consecutive days of nicotine-sucrose pairing. Our findings extend previous reports to a contingent model of nicotine self-administration and show that p-dmCPu is involved in associative learning with nicotine stimulus using a paradigm where rats voluntarily self-administer nicotine infusions that are paired with access to sucrose-a paradigm that closely resembles learning processes observed in humans.


Subject(s)
Appetitive Behavior , Association Learning , Caudate Nucleus , Central Nervous System Agents/administration & dosage , Goals , Nicotine/administration & dosage , Putamen , Animals , Appetitive Behavior/drug effects , Appetitive Behavior/physiology , Association Learning/drug effects , Association Learning/physiology , Caudate Nucleus/drug effects , Caudate Nucleus/physiopathology , Male , Putamen/drug effects , Putamen/physiopathology , Rats , Rats, Sprague-Dawley , Self Administration , Sucrose/administration & dosage , Sweetening Agents/administration & dosage
8.
Folia Neuropathol ; 59(2): 113-120, 2021.
Article in English | MEDLINE | ID: mdl-34284539

ABSTRACT

The majority of COVID-19 cases are only mildly or moderately symptomatic, but in some patients excessive inflammatory response becomes the dominant factor of disease progression to the advanced stage, with high mortality. Treatment with anti-inflammatory drugs either does not prevent disease progression (non-steroidal anti-inflammatory drugs [NSAIDs], colchicine), or is recommended only at the advanced disease stage (dexamethasone). Fluvoxamine and amantadine are drugs used to treat neurological and psychiatric diseases. Fluvoxamine is a selective serotonin uptake inhibitor, whereas amantadine is an old antiviral variably influencing brain neurotransmitter systems, and repurposed to Parkinson's disease. Both drugs are agonists of sigma-1 receptors located in the endoplasmic reticulum, which effect seems responsible for their anti-inflammatory activity. Moreover, amantadine was found to dampen the expression of cathepsin-L, a lysosomal enzyme implicated in SARS-CoV-2 virus entry to target cells. In two small controlled clinical trials, early treatment of SARS-CoV-2-infected persons with fluvoxamine fully prevented COVID-19 symptoms. Anecdotal evidence shows that amantadine may be similarly effective. Both drugs are easily available, inexpensive and have favorable safety profiles. Clinical trials evaluating their efficacy as much-needed post-exposure prophylaxis and early treatment of COVID-19 are ongoing.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Antiviral Agents/administration & dosage , COVID-19 Drug Treatment , Central Nervous System Agents/administration & dosage , Selective Serotonin Reuptake Inhibitors/administration & dosage , COVID-19/diagnosis , COVID-19/immunology , Humans , Treatment Outcome
10.
J Zoo Wildl Med ; 52(1): 287-294, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33827187

ABSTRACT

This study investigated the use of a fixed-dose combination of 30 mg/ml butorphanol, 12 mg/ml azaperone, and 12 mg/ml medetomidine for the standing sedation of captive African elephants (Loxodonta africana). In total, seven females (mean age 19.6 yr; range 6-31 yr) and six males (mean age 33.5 yr; range 9-35 yr) were sedated. The estimated dose was 0.0005 ± 0.0001 ml/kg and 0.006 ± 0.001 ml/cm shoulder height, which resulted in a dose of 0.016 ± 0.002 mg/kg or 0.19 ± 0.04 mg/cm shoulder height butorphanol, 0.006 ± 0.0008 mg/ kg or 0.076 ± 0.015 mg/cm shoulder height azaperone, and 0.006 ± 0.0008 mg/kg or 0.076 ± 0.015 mg/cm medetomidine. First signs of sedation were observed within 3-10 min (mean 6 ± 2 min) after darting, and monitoring of the animals started on average at 24 ± 9 min after darting. No bradycardia was observed in any of the elephants (mean heart rate 40.0 ± 6.55 beats/min), although all the animals were mildly hypotensive (mean blood pressure 118.5/86 [94.5]). Rectal temperatures fell within acceptable ranges, and respiratory parameters were stable in all the animals throughout sedation and fell within the standard ranges reported for conscious, standing elephants. Only one elephant had clinically significant hypoxemia characterized by a partial pressure of oxygen (PaO2) < 60 mm Hg. This elephant was also hypercapnic (PaCO2 > 50 mm Hg), although pH and peripheral capillary oxygen saturation fell within acceptable ranges. None of the elephants reacted to moderately painful stimuli while sedated. The combination was reversed with intramuscular injections of naltrexone (1 mg for every 1 mg butorphanol) and atipamezole (5 mg for every 1 mg medetomidine). Recovery was smooth and calm in all the animals. Time from injection of the reversals until the first signs of recovery was 4.6 ± 2.01 min (range 1-8 min).


Subject(s)
Azaperone/administration & dosage , Butorphanol/administration & dosage , Central Nervous System Agents/administration & dosage , Conscious Sedation/veterinary , Elephants/physiology , Medetomidine/administration & dosage , Analgesics, Opioid/administration & dosage , Animals , Drug Combinations , Female , Hypnotics and Sedatives/administration & dosage , Male , Naltrexone/administration & dosage , Narcotic Antagonists/administration & dosage
11.
Am J Physiol Renal Physiol ; 320(5): F963-F971, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33843270

ABSTRACT

Psychotropic drugs may be associated with hyponatremia, but an understanding of how they induce water retention in the kidney remains elusive. Previous studies have postulated that they may increase vasopressin production in the hypothalamus without supporting evidence. In this study, we investigated the possibility of drug-induced nephrogenic syndrome of inappropriate antidiuresis using haloperidol, sertraline, and carbamazepine. Haloperidol, sertraline, or carbamazepine were treated in inner medullary collecting duct (IMCD) suspensions and primary cultured IMCD cells prepared from male Sprague-Dawley rats. The responses of intracellular cAMP production, aquaporin-2 (AQP2) protein expression and localization, vasopressin-2 receptor (V2R) and AQP2 mRNA, and cAMP-responsive element-binding protein (CREB) were tested with and without tolvaptan and the protein kinase A (PKA) inhibitors H89 and Rp-cAMPS. In IMCD suspensions, cAMP production was increased by haloperidol, sertraline, or carbamazepine and was relieved by tolvaptan cotreatment. In primary cultured IMCD cells, haloperidol, sertraline, or carbamazepine treatment increased total AQP2 and decreased phosphorylated Ser261-AQP2 protein expression. Notably, these responses were reversed by cotreatment with tolvaptan or a PKA inhibitor. AQP2 membrane trafficking was induced by haloperidol, sertraline, or carbamazepine and was also blocked by cotreatment with tolvaptan or a PKA inhibitor. Furthermore, upregulation of V2R and AQP2 mRNA and phosphorylated CREB was induced by haloperidol, sertraline, or carbamazepine and was blocked by tolvaptan cotreatment. We conclude that, in the rat IMCD, psychotropic drugs upregulate AQP2 via V2R-cAMP-PKA signaling in the absence of vasopressin stimulation. The vasopressin-like action on the kidney appears to accelerate AQP2 transcription and dephosphorylate AQP2 at Ser261.NEW & NOTEWORTHY It is unclear whether antipsychotic drugs can retain water in the kidney in the absence of vasopressin. This study demonstrates that haloperidol, sertraline, and carbamazepine can produce nephrogenic syndrome of inappropriate antidiuresis because they directly upregulate vasopressin-2 receptor and aquaporin-2 (AQP2) via cAMP/PKA signaling. We showed that, in addition to AQP2 trafficking, AQP2 protein abundance was rapidly increased by treatment with antipsychotic drugs in association with dephosphorylation of AQP2 at Ser261 and accelerated AQP2 transcription.


Subject(s)
Aquaporin 2/metabolism , Central Nervous System Agents/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Kidney Tubules, Collecting/cytology , Receptors, Vasopressin/metabolism , Animals , Carbamazepine/administration & dosage , Carbamazepine/pharmacology , Central Nervous System Agents/administration & dosage , Cyclic AMP/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Drug Therapy, Combination , Gene Expression Regulation/drug effects , Haloperidol/administration & dosage , Haloperidol/pharmacology , Male , Phosphorylation , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Vasopressin/genetics , Sertraline/administration & dosage , Sertraline/pharmacology , Vasopressins/administration & dosage , Vasopressins/pharmacology
12.
Pharm Dev Technol ; 26(6): 617-633, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33882793

ABSTRACT

Despite a relatively low prevalence of primary brain tumors, they continuously attract scientific interest because of the complexity of their treatment due to their location behind the blood-brain barrier. The main challenge in treatment of brain tumors is not the efficacy of the drugs, per se, but the low efficiency of drug delivery to malignant cells. At the core of the problem is the complex structure of the blood-brain barrier. Nowadays, there is evidence supporting the claim that bile acids have the ability to cross the blood-brain barrier. That ability can be exploited by taking a part in novel drug carrier designs. Bile acids represent a drug carrier system as a part of a mixed micelle composition, bilosomes and conjugates with various drugs. This review discusses the current knowledge related to bile acid molecules as drug penetration modifying agents, with the focus on central nervous system antitumor drug delivery.


Subject(s)
Antineoplastic Agents/metabolism , Bile Acids and Salts/metabolism , Blood-Brain Barrier/metabolism , Central Nervous System Agents/metabolism , Drug Delivery Systems/methods , Neoplasms/metabolism , Animals , Antineoplastic Agents/administration & dosage , Bile Acids and Salts/administration & dosage , Biological Transport/drug effects , Biological Transport/physiology , Blood-Brain Barrier/drug effects , Central Nervous System Agents/administration & dosage , Drug Carriers/administration & dosage , Drug Carriers/metabolism , Humans , Neoplasms/drug therapy
13.
Neurosci Lett ; 755: 135911, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33892003

ABSTRACT

Amongst the most important discoveries in ALS pathobiology are the works demonstrating that multiple cell types contribute to disease onset and progression. However, a significant limitation in ALS research is the inability to obtain tissues from ALS patient brain and spinal cord during the course of the disease. In vivo modeling has provided insights into the role of these cell subtypes in disease onset and progression. However, in vivo models also have shortcomings, including the reliance on a limited number of models based upon hereditary forms of the disease. Therefore, using human induced pluripotent stem cells (iPSC) reprogrammed from somatic cells of ALS patients, with both hereditary and sporadic forms of the disease, and differentiated into cell subtypes of both the central nervous system (CNS) and peripheral nervous system (PNS), have become powerful complementary tools for investigating basic mechanisms of disease as well as a platform for drug discovery. Motor neuron and other neuron subtypes, as well as non-neuronal cells have been differentiated from human iPSC and studied for their potential contributions to ALS pathobiology. As iPSC technologies have advanced, 3D modeling with multicellular systems organised in microfluidic chambers or organoids are the next step in validating the pathways and therapeutic targets already identified. Precision medicine approaches with iPSC using either traditional strategies of screening drugs that target a known pathogenic mechanism as well as "blind-to-target" drug screenings that allow for patient stratification based on drug response rather than clinical characteristics are now being employed.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , Cellular Reprogramming Techniques/methods , Induced Pluripotent Stem Cells/transplantation , Stem Cell Transplantation/methods , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cellular Reprogramming Techniques/trends , Central Nervous System Agents/administration & dosage , Coculture Techniques , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/trends , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/physiology , Organoids/cytology , Organoids/drug effects , Organoids/physiology , Stem Cell Transplantation/trends
14.
Behav Brain Res ; 408: 113294, 2021 06 25.
Article in English | MEDLINE | ID: mdl-33836167

ABSTRACT

Paracetamol (PAR) has been employed worldwide for pain and fever treatment during pregnancy and lactation. Epidemiologic studies have shown that exposure to PAR can increase the risk for developmental disorders, such as attention-deficit hyperactive disorder and autism spectrum disorder. This study aimed to investigate if gestational and lactational exposure to human-relevant doses of PAR could alter behavioural and brain oxidative stress parameters in the rat`s offspring. Wistar dams were gavaged daily with water or PAR (35 mg/kg/ or 350 mg/kg) during gestational day 6 to weaning (postnatal day 21). Behavioural assessments occurred at post-natal days 10 (nest seeking test), 27 (behavioural stereotypy) and 28 (three chamber sociability test and open field). Concentration of advanced oxidation protein products (AOPP), reduced glutathione (GSH), lipid hydroperoxides (LOOH) and activity of superoxide dismutase (SOD) were estimate in prefrontal cortex, hippocampus, striatum and cerebellum of 22-day-old rats. Compared to CON animals, males exposed to PAR during pregnancy and lactation augmented apomorphine-induced stereotyped behaviour (350 mg/kg) and ambulation in open-field test (35 mg/kg). Reduced exploratory behaviour in three chamber sociability test was observed in pups exposed to PAR at 350 mg/kg in both sexes. PAR treatment decreased hippocampal GSH level and striatal SOD activity in males exposed to 35 mg/kg, suggesting the vulnerability of these areas in PAR-induced developmental neurotoxicity. Findings suggest PAR use during pregnancy and lactation as a potential risk factor for neurodevelopmental disorders with males being more susceptible.


Subject(s)
Acetaminophen/pharmacology , Behavior, Animal/drug effects , Central Nervous System Agents/pharmacology , Exploratory Behavior/drug effects , Hippocampus/drug effects , Neurodevelopmental Disorders/chemically induced , Oxidative Stress/drug effects , Prenatal Exposure Delayed Effects/chemically induced , Acetaminophen/administration & dosage , Animals , Behavior, Animal/physiology , Breast Feeding , Central Nervous System Agents/administration & dosage , Disease Models, Animal , Exploratory Behavior/physiology , Female , Hippocampus/metabolism , Male , Neurodevelopmental Disorders/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Rats , Rats, Wistar , Sex Factors
15.
Clin Ter ; 172(2): 116-118, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33763670

ABSTRACT

ABSTRACT: Over the last few years reports have indicated an increase in the number, type and availability of new psychoactive substances belonging to the benzodiazepine class. These molecules may pose high risks to users, since the majority have never undergone clinical trials or tests so their pharmacology and toxicology is largely unknown. However the new drug scenario emerging from the COVID-19 global pandemic seems to play a role in increasing the diversion of prescribed benzodiazepines and Z-drug. A brief presentation of this phenomenon is hereby presented. The awareness and response activities at national and international levels related to this issue should be enforced.


Subject(s)
Benzodiazepines/adverse effects , COVID-19 , Substance-Related Disorders/epidemiology , Benzodiazepines/administration & dosage , Central Nervous System Agents/administration & dosage , Central Nervous System Agents/adverse effects , Humans , Prescription Drug Diversion/trends
16.
Life Sci ; 274: 119326, 2021 Jun 01.
Article in English | MEDLINE | ID: mdl-33711385

ABSTRACT

The blood-brain barrier (BBB) is composed of a layer of endothelial cells that is interspersed with a series of tight junctions and characterized by the absence of fenestrations. The permeability of this barrier is controlled by junctions such as tight junctions and adherent junctions as well as several cells such as astrocytes, pericytes, vascular endothelial cells, neurons, microglia, and efflux transporters with relatively enhanced expression. It plays a major role in maintaining homeostasis in the brain and exerts a protective regulatory control on the influx and efflux of molecules. However, it proves to be a challenge for drug delivery strategies that target brain diseases like Dementia, Parkinson's Disease, Alzheimer's Disease, Brain Cancer or Stroke, Huntington's Disease, Lou Gehrig's Disease, etc. Conventional modes of drug delivery are invasive and have been known to contribute to a "leaky BBB", recent studies have highlighted the efficiency and relative safety of receptor-mediated drug delivery. Several receptors are exhibited on the BBB, and actively participate in nutrient uptake, and recognize specific ligands that modulate the process of endocytosis. The strategy employed in receptor-mediated drug delivery exploits this process of "tricking" the receptors into internalizing ligands that are conjugated to carrier systems like liposomes, nanoparticles, monoclonal antibodies, enzymes etc. These in turn are modified with drug molecules, therefore leading to delivery to desired target cells in brain tissue. This review comprehensively explores each of those receptors that can be modified to serve such purposes as well as the currently employed strategies that have led to increased cellular uptake and transport efficiency.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Diseases/drug therapy , Central Nervous System Agents/administration & dosage , Drug Carriers/chemistry , Drug Delivery Systems , Receptors, Cell Surface/metabolism , Animals , Biological Transport , Blood-Brain Barrier/metabolism , Brain Diseases/metabolism , Brain Diseases/pathology , Humans , Ligands , Permeability
17.
Expert Opin Drug Metab Toxicol ; 17(8): 923-936, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33595380

ABSTRACT

INTRODUCTION: The increasing incidence of mental illnesses and neurodegenerative diseases results in a high demand for drugs targeting the central nervous system (CNS). These drugs easily reach the CNS, have a high affinity for CNS targets, and are prone to cause seizures as an adverse drug reaction. Current seizure liability assessment heavily depends on in vivo or ex vivo animal models and is therefore ethically debated, labor intensive, expensive, and not always predictive for human risk. AREAS COVERED: The demand for CNS drugs urges the development of alternative safety assessment strategies. Yet, the complexity of the CNS hampers reliable detection of compound-induced seizures. This review provides an overview of the requirements of in vitro seizure liability assays and highlights recent advances, including micro-electrode array (MEA) recordings using rodent and human cell models. EXPERT OPINION: Successful and cost-effective replacement of in vivo and ex vivo models for seizure liability screening can reduce animal use for drug development, while increasing the predictive value of the assays, particularly if human cell models are used. However, these novel test strategies require further validation and standardization as well as additional refinements to better mimic the human in vivo situation and increase their predictive value.


Subject(s)
Central Nervous System Agents/adverse effects , Models, Biological , Seizures/chemically induced , Animals , Central Nervous System Agents/administration & dosage , Cost-Benefit Analysis , Drug Development/methods , Humans , Microelectrodes , Predictive Value of Tests , Seizures/diagnosis
18.
Adv Drug Deliv Rev ; 171: 332-351, 2021 04.
Article in English | MEDLINE | ID: mdl-33497734

ABSTRACT

A variety of neuroprotectants have shown promise in treating ischemic stroke, yet their delivery to the brain remains a challenge. The endothelial cells lining the blood-brain barrier (BBB) are emerging as a dynamic factor in the response to neurological injury and disease, and the endothelial-neuronal matrix coupling is fundamentally neuroprotective. In this review, we discuss approaches that target the endothelium for drug delivery both across the BBB and to the BBB as a viable strategy to facilitate neuroprotective effects, using the example of brain-derived neurotrophic factor (BDNF). We highlight the advances in cell-derived extracellular vesicles (EVs) used for CNS targeting and drug delivery. We also discuss the potential of engineered EVs as a potent strategy to deliver BDNF or other drug candidates to the ischemic brain, particularly when coupled with internal components like mitochondria that may increase cellular energetics in injured endothelial cells.


Subject(s)
Blood-Brain Barrier/metabolism , Drug Delivery Systems , Stroke/drug therapy , Animals , Brain-Derived Neurotrophic Factor/administration & dosage , Central Nervous System Agents/administration & dosage , Extracellular Vesicles , Humans , Nucleic Acids/administration & dosage , Stroke/metabolism
19.
Curr Drug Metab ; 22(4): 251-262, 2021.
Article in English | MEDLINE | ID: mdl-33234099

ABSTRACT

Impressive research steps have been taken for the treatment of neurological disorders in the last few decades. Still, effective treatments of brain related disorders are very less due to problems associated with crossing the blood-brain barrier (BBB), non-specific therapies, and delay in functional recovery of the central nervous system (CNS) after treatment. Striving for novel treatment options for neurological disorders, nanotechnology- derived materials, and devices have gained ground due to inherent features of derivatization/encapsulation with drugs as per the neurological ailments and pharmacological targets. Facile developments/syntheses of the nanomaterials-drug conjugates have also been the driving force for researchers to get into this field. Moreover, the tunable size and hydro/lipophilicity of these nanomaterials are the added advantages that make these materials more acceptable for CNS disorders. These nano-neurotherapeutics (NNTs) systems provide the platform for diagnosis, theranostics, treatments, restoration of CNS disorders, and encourage the translation of NNTs from "bench to bedside". Still, these techniques are in the primary stages of medical development. This review describes the latest advancements and future scenarios of developmental and clinical aspects of polymeric NNTs.


Subject(s)
Blood-Brain Barrier/metabolism , Central Nervous System Agents/administration & dosage , Central Nervous System Diseases/drug therapy , Nanoparticle Drug Delivery System/pharmacology , Theranostic Nanomedicine/trends , Animals , Clinical Trials as Topic , Disease Models, Animal , Humans , Nanoparticle Drug Delivery System/chemistry , Permeability/drug effects , Theranostic Nanomedicine/methods , Treatment Outcome
20.
Biochem Pharmacol ; 187: 114367, 2021 05.
Article in English | MEDLINE | ID: mdl-33333075

ABSTRACT

The initial exploration using pharmacological tools of the role of adenosine receptors in the brain, concluded that adenosine released as such acted on A1R to inhibit excitability and glutamate release from principal neurons throughout the brain and that adenosine A2A receptors (A2AR) were striatal-'specific' receptors controlling dopamine D2R. This indicted A1R as potential controllers of neurodegeneration and A2AR of psychiatric conditions. Global knockout of these two receptors questioned the key role of A1R and instead identified extra-striatal A2AR as robust controllers of neurodegeneration. Furthermore, transgenic lines with altered metabolic sources of adenosine revealed a coupling of ATP-derived adenosine to activate A2AR and a role of A1R as a hurdle to initiate neurodegeneration. Additionally, cell-selective knockout of A2AR unveiled the different roles of A2AR in different cell types (neurons/astrocytes) in different portions of the striatal circuits (dorsal versus lateral) and in different brain areas (hippocampus/striatum). Finally, a new transgenic mouse line with deletion of all adenosine receptors seems to indicate a major allostatic rather than homeostatic role of adenosine and may allow isolating P2R-mediated responses to unravel their role in the brain, a goal close to heart of Geoffrey Burnstock, to whom we affectionately dedicate this review.


Subject(s)
Adenosine/administration & dosage , Adenosine/metabolism , Brain/metabolism , Central Nervous System Agents/administration & dosage , Central Nervous System Agents/metabolism , Receptors, Purinergic P1/metabolism , Animals , Brain/drug effects , Mice , Mice, Knockout , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...