Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.310
Filter
1.
Front Immunol ; 15: 1423263, 2024.
Article in English | MEDLINE | ID: mdl-39224601

ABSTRACT

Combination antiretroviral therapy (cART) has dramatically reduced mortality in people with human immunodeficiency virus (HIV), but it does not completely eradicate the virus from the brain. Patients with long-term HIV-1 infection often show neurocognitive impairment, which severely affects the quality of life of those infected. Methamphetamine (METH) users are at a significantly higher risk of contracting HIV-1 through behaviors such as engaging in high-risk sex or sharing needles, which can lead to transmission of the virus. In addition, HIV-1-infected individuals who abuse METH exhibit higher viral loads and more severe cognitive dysfunction, suggesting that METH exacerbates the neurotoxicity associated with HIV-1. Therefore, this review focuses on various mechanisms underlying METH and HIV-1 infection co-induced neurotoxicity and existing interventions targeting the sigma 1 receptor, dopamine transporter protein, and other relevant targets are explored. The findings of this review are envisaged to systematically establish a theoretical framework for METH abuse and HIV-1 infection co-induced neurotoxicity, and to suggest novel clinical treatment targets.


Subject(s)
HIV Infections , HIV-1 , Methamphetamine , Humans , Methamphetamine/adverse effects , HIV Infections/drug therapy , HIV Infections/complications , HIV-1/drug effects , Neurotoxicity Syndromes/etiology , Animals , Amphetamine-Related Disorders/complications , Amphetamine-Related Disorders/therapy , Sigma-1 Receptor , Dopamine Plasma Membrane Transport Proteins/metabolism
2.
J Hematol Oncol ; 17(1): 61, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107847

ABSTRACT

Autologous anti-CD19 chimeric antigen receptor (CAR) T cells are now used in routine practice for relapsed/refractory (R/R) large B-cell lymphoma (LBCL). Severe (grade ≥ 3) cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity (ICANS) are still the most concerning acute toxicities leading to frequent intensive care unit (ICU) admission, prolonging hospitalization, and adding significant cost to treatment. We report on the incidence of CRS and ICANS and the outcomes in a large cohort of 925 patients with LBCL treated with axicabtagene ciloleucel (axi-cel) or tisagenlecleucel (tisa-cel) in France based on patient data captured through the DESCAR-T registry. CRS of any grade occurred in 778 patients (84.1%), with 74 patients (8.0%) with grade 3 CRS or higher, while ICANS of any grade occurred in 375 patients (40.5%), with 112 patients (12.1%) with grade ≥ 3 ICANS. Based on the parameters selected by multivariable analyses, two independent prognostic scoring systems (PSS) were derived, one for grade ≥ 3 CRS and one for grade ≥ 3 ICANS. CRS-PSS included bulky disease, a platelet count < 150 G/L, a C-reactive protein (CRP) level > 30 mg/L and no bridging therapy or stable or progressive disease (SD/PD) after bridging. Patients with a CRS-PSS score > 2 had significantly higher risk to develop grade ≥ 3 CRS. ICANS-PSS included female sex, low level of platelets (< 150 G/L), use of axi-cel and no bridging therapy or SD/PD after bridging. Patients with a CRS-PSS score > 2 had significantly higher risk to develop grade ≥ 3 ICANS. Both scores were externally validated in international cohorts of patients treated with tisa-cel or axi-cel.


Subject(s)
Antigens, CD19 , Cytokine Release Syndrome , Immunotherapy, Adoptive , Lymphoma, Large B-Cell, Diffuse , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Male , Female , Middle Aged , Antigens, CD19/immunology , Prognosis , Lymphoma, Large B-Cell, Diffuse/therapy , Lymphoma, Large B-Cell, Diffuse/immunology , Cytokine Release Syndrome/etiology , Aged , Adult , Neurotoxicity Syndromes/etiology , Biological Products/therapeutic use , Biological Products/adverse effects , France , Aged, 80 and over , Receptors, Antigen, T-Cell
3.
Sci Rep ; 14(1): 19341, 2024 08 20.
Article in English | MEDLINE | ID: mdl-39164364

ABSTRACT

Peripheral neurotoxicity is a dose-limiting adverse reaction of primary frontline chemotherapeutic agents, including vincristine. Neuropathy can be so disabling that patients drop out of potentially curative therapy, negatively impacting cancer prognosis. The hallmark of vincristine neurotoxicity is axonopathy, yet its underpinning mechanisms remain uncertain. We developed a comprehensive drug discovery platform to identify neuroprotective agents against vincristine-induced neurotoxicity. Among the hits identified, SIN-1-an active metabolite of molsidomine-prevents vincristine-induced axonopathy in both motor and sensory neurons without compromising vincristine anticancer efficacy. Mechanistically, we found that SIN-1's neuroprotective effect is mediated by activating soluble guanylyl cyclase. We modeled vincristine-induced peripheral neurotoxicity in rats to determine molsidomine therapeutic potential in vivo. Vincristine administration induced severe nerve damage and mechanical hypersensitivity that were attenuated by concomitant treatment with molsidomine. This study provides evidence of the neuroprotective properties of molsidomine and warrants further investigations of this drug as a therapy for vincristine-induced peripheral neurotoxicity.


Subject(s)
Molsidomine , Neuroprotective Agents , Soluble Guanylyl Cyclase , Vincristine , Vincristine/adverse effects , Vincristine/pharmacology , Vincristine/toxicity , Animals , Neuroprotective Agents/pharmacology , Rats , Soluble Guanylyl Cyclase/metabolism , Molsidomine/pharmacology , Molsidomine/analogs & derivatives , Humans , Male , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/prevention & control , Peripheral Nervous System Diseases/drug therapy , Rats, Sprague-Dawley , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/prevention & control , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/etiology
4.
Int J Mol Sci ; 25(15)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39125900

ABSTRACT

The effects of exposure to environmental pollutants on neurological processes are of increasing concern due to their potential to induce oxidative stress and neurotoxicity. Considering that many industries are currently using different types of plastics as raw materials, packaging, or distribution pipes, microplastics (MPs) have become one of the biggest threats to the environment and human health. These consequences have led to the need to raise the awareness regarding MPs negative neurological effects and implication in neuropsychiatric pathologies, such as schizophrenia. The study aims to use three zebrafish models of schizophrenia obtained by exposure to ketamine (Ket), methionine (Met), and their combination to investigate the effects of MP exposure on various nervous system structures and the possible interactions with oxidative stress. The results showed that MPs can interact with ketamine and methionine, increasing the severity and frequency of optic tectum lesions, while co-exposure (MP+Met+Ket) resulted in attenuated effects. Regarding oxidative status, we found that all exposure formulations led to oxidative stress, changes in antioxidant defense mechanisms, or compensatory responses to oxidative damage. Met exposure induced structural changes such as necrosis and edema, while paradoxically activating periventricular cell proliferation. Taken together, these findings highlight the complex interplay between environmental pollutants and neurotoxicants in modulating neurotoxicity.


Subject(s)
Brain , Disease Models, Animal , Microplastics , Oxidative Stress , Schizophrenia , Zebrafish , Zebrafish/metabolism , Animals , Oxidative Stress/drug effects , Microplastics/toxicity , Schizophrenia/metabolism , Schizophrenia/chemically induced , Schizophrenia/pathology , Schizophrenia/etiology , Brain/metabolism , Brain/drug effects , Brain/pathology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/pathology , Ketamine/adverse effects , Ketamine/toxicity , Methionine/metabolism , Immunohistochemistry
5.
Cell Biol Toxicol ; 40(1): 63, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39093513

ABSTRACT

Anesthetic-induced developmental neurotoxicity (AIDN) can arise due to various factors, among which aberrant nerve cell death is a prominent risk factor. Animal studies have reported that repeated or prolonged anesthetic exposure can cause significant neuroapoptosis in the developing brain. Lately, non-apoptotic programmed cell deaths (PCDs), characterized by inflammation and oxidative stress, have gained increasing attention. Substantial evidence suggests that non-apoptotic PCDs are essential for neuronal cell death in AIDN compared to apoptosis. This article examines relevant publications in the PubMed database until April 2024. Only original articles in English that investigated the potential manifestations of non-apoptotic PCD in AIDN were analysed. Specifically, it investigates necroptosis, pyroptosis, ferroptosis, and parthanatos, elucidating the signaling mechanisms associated with each form. Furthermore, this study explores the potential relevance of these non-apoptotic PCDs pathways to the pathological mechanisms underlying AIDN, drawing upon their distinctive characteristics. Despite the considerable challenges involved in translating fundamental scientific knowledge into clinical therapeutic interventions, this comprehensive review offers a theoretical foundation for developing innovative preventive and treatment strategies targeting non-apoptotic PCDs in the context of AIDN.


Subject(s)
Anesthetics , Apoptosis , Neurotoxicity Syndromes , Humans , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/etiology , Animals , Anesthetics/adverse effects , Anesthetics/toxicity , Anesthetics/pharmacology , Apoptosis/drug effects , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Pyroptosis/drug effects , Oxidative Stress/drug effects , Necroptosis/drug effects , Brain/drug effects , Brain/pathology , Brain/growth & development , Ferroptosis/drug effects , Signal Transduction/drug effects
6.
Sci Rep ; 14(1): 18548, 2024 08 09.
Article in English | MEDLINE | ID: mdl-39122917

ABSTRACT

This study aimed to elucidate the incidence and characteristics of neurotoxicity in patients receiving methotrexate (MTX) treatment. A retrospective analysis was performed using data from the electronic cohort database spanning from January 1990 to December 2021. This review focused on patients who manifested neurotoxic symptoms post-MTX therapy, excluding patients with peripheral neuropathy. Of the 498 individuals who received MTX, 26 (5.22%) exhibited neurotoxicity. Pediatric patients (< 18 years) accounted for 18 cases (7.44%), whereas adults (> 18 years) comprised eight cases (3.13%). The median onset age was 11 years (range 4-15) in the pediatric cohort and 39.5 years (range 19-67) in the adult cohort. A predominant male predisposition was noted (21 patients, 80.77%). The majority of patients (21, 80.77%) experienced neurotoxic effects following multiple MTX administrations. Modes of MTX delivery included intrathecal (37.0%), intravenous (22.2%), and combined routes (40.7%). Clinical presentations were predominantly encephalopathy (69.2%), followed by encephalomyelopathy (15.4%), myelopathy (11.5%), and polyradiculopathy (3.8%). Fourteen patients recovered (53.85%). Risk factors were male sex, pediatric age (particularly above 10 years), and administration route (intrathecal in adults and intravenous in pediatrics). Although infrequent, MTX-related neurotoxicity has a substantial impact on patient prognosis, with potential development following even a single dose. Its radiological resemblance to diverse neuropathologies, such as cerebral infarction and subacute combined degeneration, necessitates vigilant diagnostic scrutiny.


Subject(s)
Methotrexate , Neurotoxicity Syndromes , Humans , Methotrexate/adverse effects , Methotrexate/administration & dosage , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/epidemiology , Male , Female , Adult , Retrospective Studies , Child , Adolescent , Child, Preschool , Aged , Middle Aged , Young Adult , Incidence
8.
Sci Rep ; 14(1): 19075, 2024 08 17.
Article in English | MEDLINE | ID: mdl-39154055

ABSTRACT

Human induced pluripotent stem cell-derived sensory neuron (iPSC-dSN) models are a valuable resource for the study of neurotoxicity but are affected by poor replicability and reproducibility, often due to a lack of optimization. Here, we identify experimental factors related to culture conditions that substantially impact cellular drug response in vitro and determine optimal conditions for improved replicability and reproducibility. Treatment duration and cell seeding density were both found to be significant factors, while cell line differences also contributed to variation. A replicable dose-response in viability was demonstrated after 48-h exposure to docetaxel or paclitaxel. Additionally, a replicable dose-dependent reduction in neurite outgrowth was demonstrated, demonstrating the applicability of the model for the examination of additional phenotypes. Overall, we have established an optimized iPSC-dSN model for the study of taxane-induced neurotoxicity.


Subject(s)
Cell Survival , Induced Pluripotent Stem Cells , Sensory Receptor Cells , Taxoids , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/cytology , Taxoids/pharmacology , Sensory Receptor Cells/drug effects , Cell Survival/drug effects , Docetaxel/pharmacology , Neurotoxicity Syndromes/etiology , Bridged-Ring Compounds/pharmacology , Cell Differentiation/drug effects , Paclitaxel/pharmacology , Paclitaxel/toxicity , Cell Line , Cells, Cultured
9.
Biomolecules ; 14(8)2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39199372

ABSTRACT

Pregnancy is a particularly vulnerable period for the growing fetus, when exposure to toxic agents, especially in the early phases, can decisively harm embryo development and compromise the future health of the newborn. The inclusion of various chemical substances in personal care products (PCPs) and cosmetic formulations can be associated with disruption and damage to the nervous system. Microplastics, benzophenones, parabens, phthalates and metals are among the most common chemical substances found in cosmetics that have been shown to induce neurotoxic mechanisms. Although cosmetic neurotoxin exposure is believed to be minimal, different exposure scenarios of cosmetics suggest that these neurotoxins remain a threat. Special attention should be paid to early exposure in the first weeks of gestation, when critical processes, like the migration and proliferation of the neural crest derived cells, start to form the ENS. Importantly, cosmetic neurotoxins can cross the placental barrier and affect the future embryo, but they are also secreted in breast milk, so babies remain exposed for longer periods, even after birth. In this review, we explore how neurotoxins contained in cosmetics and PCPs may have a role in the pathogenesis of various neurodevelopmental disorders and neurodegenerative diseases and, therefore, also in congenital enteric aganglionosis as well as in postnatal motility disorders. Understanding the mechanisms of these chemicals used in cosmetic formulations and their role in neurotoxicity is crucial to determining the safety of use for cosmetic products during pregnancy.


Subject(s)
Cosmetics , Humans , Female , Pregnancy , Cosmetics/adverse effects , Neurotoxins/toxicity , Neurotoxicity Syndromes/etiology , Phthalic Acids/toxicity , Animals
10.
BMC Infect Dis ; 24(1): 862, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39187812

ABSTRACT

BACKGROUND: The study aimed to compare polymyxin B with colistimethate sodium (CMS) regarding neurotoxicity, nephrotoxicity and 30-day mortality in patients with MDR Gram-negatives. METHODS: All adult patients who received polymyxin B or CMS for at least 24 h for the treatment of MDR microorganisms were evaluated retrospectively. RESULTS: Among 413 initially screened patients, 147 patients who were conscious and able to express their symptoms were included in the neurotoxicity analysis. 13 of 77 patients with polymyxin B and 1 of 70 with CMS had neurotoxic adverse events, mainly paresthesias. All events were reversible after drug discontinuation. Among 290 patients included in nephrotoxicity analysis, the incidence of acute kidney injury (AKI) was 44.7% and 40.0% for polymyxin B and CMS, respectively (p = 0.425). AKI occurred two days earlier with colistin than polymyxin B without statistical significance (median (IQR): 5 (3-11) vs. 7 (3-12), respectively, p = 0.701). Polymyxin therapy was withdrawn in 41.1% of patients after AKI occurred and CMS was more frequently withdrawn than polymyxin B (p = 0.025). AKI was reversible in 91.6% of patients with CMS and 79% with polymyxin B after the drug withdrawal. Older age, higher baseline serum creatinine and the use of at least two nephrotoxic drugs were independent factors associated with AKI (OR 1.05, p < 0.001; OR 2.99, p = 0.022 and OR 2.45, p = 0.006, respectively). Septic shock, mechanical ventilation, presence of a central venous catheter and Charlson comorbidity index (OR 2.13, p = 0.004; OR 3.37, p < 0.001; OR 2.47, p = 0.004 and OR 1.21, p p < 0.001, respectively) were the independent predictors of mortality. The type of polymyxin was not related to mortality. CONCLUSIONS: Neurotoxicity is a relatively common adverse event that leads to drug withdrawal during polymyxins, particularly polymyxin B. Nephrotoxicity is very common during polymyxin therapy and the two polymyxins display similar nephrotoxic events with high reversibility rates after drug withdrawal. Close monitoring of AKI is crucial during polymyxin therapy, particularly, for elderly patients, patients who have high baseline creatinine, and using other nephrotoxic drugs.


Subject(s)
Acute Kidney Injury , Anti-Bacterial Agents , Colistin , Polymyxin B , Humans , Colistin/adverse effects , Colistin/analogs & derivatives , Polymyxin B/adverse effects , Polymyxin B/therapeutic use , Male , Female , Middle Aged , Retrospective Studies , Aged , Anti-Bacterial Agents/adverse effects , Acute Kidney Injury/chemically induced , Gram-Negative Bacterial Infections/drug therapy , Adult , Drug Resistance, Multiple, Bacterial , Aged, 80 and over , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/epidemiology
11.
ACS Nano ; 18(35): 24044-24059, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39158845

ABSTRACT

Oral ingestion is the primary route for human exposure to nanoplastics, making the gastrointestinal tract one of the first and most impacted organs. Given the presence of the gut-brain axis, a crucial concern arises regarding the potential impact of intestinal damage on the neurotoxic effects of nanoplastics (NPs). The intricate mechanisms underlying NP-induced neurotoxicity through the microbiome-gut-brain axis necessitate further investigation. To address this, we used mice specifically engineered with nuclear factor erythroid-derived 2-related factor 2 (Nrf2) deficiency in their intestines, a strain whose intestines are particularly susceptible to polystyrene NPs (PS-NPs). We conducted a 28-day repeated-dose oral toxicity study with 2.5 and 250 mg/kg of 50 nm PS-NPs in these mice. Our study delineated how PS-NP exposure caused gut microbiota dysbiosis, characterized by Mycoplasma and Coriobacteriaceae proliferation, resulting in increased levels of interleukin 17C (IL-17C) production in the intestines. The surplus IL-17C permeated the brain via the bloodstream, triggering inflammation and brain damage. Our investigation elucidated a direct correlation between intestinal health and neurological outcomes in the context of PS-NP exposure. Susceptible mice with fragile guts exhibited heightened neurotoxicity induced by PS-NPs. This phenomenon was attributed to the elevated abundance of microbiota associated with IL-17C production in the intestines of these mice, such as Mesorhizobium and Lwoffii, provoked by PS-NPs. Neurotoxicity was alleviated by in vivo treatment with anti-IL-17C-neutralizing antibodies or antibiotics. These findings advanced our comprehension of the regulatory mechanisms governing the gut-brain axis in PS-NP-induced neurotoxicity and underscored the critical importance of maintaining intestinal health to mitigate the neurotoxic effects of PS-NPs.


Subject(s)
Brain , NF-E2-Related Factor 2 , Polystyrenes , Animals , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , Mice , Polystyrenes/chemistry , Polystyrenes/toxicity , Brain/drug effects , Brain/metabolism , Brain/pathology , Gastrointestinal Microbiome/drug effects , Nanoparticles/chemistry , Microplastics/toxicity , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice, Inbred C57BL , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/pathology
12.
Intensive Care Med ; 50(9): 1459-1469, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39172238

ABSTRACT

The use of chimeric antigen receptor T (CAR-T) cells is a significant therapeutic improvement increasing the prognosis for patients with a variety of hematological malignancies. However, this therapy has also sometimes life-threatening, complications. Therefore, knowledge of the treatment and management of these complications, especially in treatment centers and intensive care units, respectively, is of outstanding importance. This review provides recommendations for the diagnosis, management, and treatment of CAR-T cell-associated complications such as cytokine release syndrome, immune effector cell associated neurotoxicity syndrome, hematotoxicity, hypogammaglobulinemia, and CAR-T cell-induced pseudo-progression amongst others for physicians treating patients with CAR-T cell-associated complications and intensivists.


Subject(s)
Cytokine Release Syndrome , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/therapeutic use , Receptors, Chimeric Antigen/immunology , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/therapy , Hematologic Neoplasms/therapy , Hematologic Neoplasms/immunology , Neurotoxicity Syndromes/etiology
13.
Expert Opin Drug Metab Toxicol ; 20(7): 629-646, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38984683

ABSTRACT

AREAS COVERED: This paper outlines the selection of NAMs, including in vitro assays using primary rat cortical neurons, zebrafish embryos, and Caenorhabditis elegans. These assays aim to assess neurotoxic endpoints such as neuronal activity and behavioral responses. Microelectrode array recordings of rat cortical neurons provide insights into the impact of botanical extracts on neuronal function, while the zebrafish embryos and C. elegans assays evaluate neurobehavioral responses. The paper also provides an account of the selection of botanical case studies based on expert judgment and existing neuroactivity/toxicity information. The proposed battery of assays will be tested with these case studies to evaluate their utility for neurotoxicity screening. EXPERT OPINION: The complexity of botanicals necessitates the use of multiple NAMs for effective neurotoxicity screening. This paper discusses the evaluation of methodologies to develop a robust framework for evaluating botanical safety, including complex neuronal models and key neurodevelopmental process assays. It aims to establish a comprehensive screening framework.


Subject(s)
Caenorhabditis elegans , Neurons , Neurotoxicity Syndromes , Toxicity Tests , Zebrafish , Animals , Neurons/drug effects , Caenorhabditis elegans/drug effects , Rats , Neurotoxicity Syndromes/diagnosis , Neurotoxicity Syndromes/etiology , Humans , Toxicity Tests/methods , Plant Extracts/adverse effects , Plant Extracts/pharmacology , Plant Extracts/administration & dosage , Drug Evaluation, Preclinical/methods , Plant Preparations/adverse effects , Plant Preparations/toxicity , Plant Preparations/pharmacology , Embryo, Nonmammalian/drug effects
14.
FASEB J ; 38(14): e23793, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39003634

ABSTRACT

Sevoflurane, as a commonly used inhaled anesthetic for pediatric patients, has been reported that multiple sevoflurane exposures are associated with a greater risk of developing neurocognitive disorder. N6-Methyladenosine (m6A), as the most common mRNA modification in eukaryotes, has emerged as a crucial regulator of brain function in processes involving synaptic plasticity, learning and memory, and neurodevelopment. Nevertheless, the relevance of m6A RNA methylation in the multiple sevoflurane exposure-induced developmental neurotoxicity remains mostly elusive. Herein, we evaluated the genome-wide m6A RNA modification and gene expression in hippocampus of mice that received with multiple sevoflurane exposures using m6A-sequencing (m6A-seq) and RNA-sequencing (RNA-seq). We discovered 19 genes with differences in the m6A methylated modification and differential expression in the hippocampus. Among these genes, we determined that a total of nine differential expressed genes may be closely associated with the occurrence of developmental neurotoxicity induced by multiple sevoflurane exposures. We further found that the alkB homolog 5 (ALKBH5), but not methyltransferase-like 3 (METTL3) and Wilms tumor 1-associated protein (WTAP), were increased in the hippocampus of mice that received with multiple sevoflurane exposures. And the IOX1, as an inhibitor of ALKBH5, significantly improved the learning and memory defects and reduced neuronal damage in the hippocampus of mice induced by multiple sevoflurane exposures. The current study revealed the role of m6A methylated modification and m6A-related regulators in sevoflurane-induced cognitive impairment, which might provide a novel insight into identifying biomarkers and therapeutic strategies for inhaled anesthetic-induced developmental neurotoxicity.


Subject(s)
Adenosine , AlkB Homolog 5, RNA Demethylase , Hippocampus , Neurotoxicity Syndromes , Sevoflurane , Sevoflurane/toxicity , Animals , Mice , AlkB Homolog 5, RNA Demethylase/metabolism , AlkB Homolog 5, RNA Demethylase/genetics , Hippocampus/metabolism , Hippocampus/drug effects , Male , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , Adenosine/analogs & derivatives , Adenosine/metabolism , Anesthetics, Inhalation/toxicity , Mice, Inbred C57BL , Methylation/drug effects , Methyltransferases/metabolism , Methyltransferases/genetics
15.
J Investig Med High Impact Case Rep ; 12: 23247096241259534, 2024.
Article in English | MEDLINE | ID: mdl-39068595

ABSTRACT

Cefepime is a fourth-generation cephalosporin with extended antimicrobial coverage. Concerns have been raised about the side effects of cefepime including myoclonus, encephalopathy, and seizures, especially when renal impairment is present. There have been reports of cases of adverse neurological consequences despite appropriate renal adjustment. Here, we present a case of a 69-year-old patient initially diagnosed with pneumonia and treated with cefepime. The patient later developed altered mental status, leading to differential diagnoses including stroke, drug overdose, or non-convulsive seizures. Following a comprehensive workup, it was determined that she had cefepime-induced encephalopathy, despite having normal kidney function, which resolved completely after discontinuing the medication. In addition, we include similar cases retrieved from PubMed up to the present date, to the best of our knowledge.


Subject(s)
Anti-Bacterial Agents , Brain Diseases , Cefepime , Intensive Care Units , Neurotoxicity Syndromes , Humans , Cefepime/adverse effects , Aged , Female , Anti-Bacterial Agents/adverse effects , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/diagnosis , Brain Diseases/chemically induced , Cephalosporins/adverse effects
17.
Toxicol Lett ; 399: 34-42, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39009234

ABSTRACT

The underlying mechanism of the aluminum (Al) on neurotoxicity remains unclear. We explored whether the impairment of hippocampal neurons induced by developmental Al exposure was associated with the m6A RNA modification in mice. In this study, the pregnant female mice were administered 4 mg/mL aluminum-lactate from gestational day (GD) 6 to postnatal day (PND) 21. On PND 21, 10 offsprings per group were euthanized by exsanguination from the abdominal aorta after deep anesthetization. The other offsprings which treated with aluminum-lactate on maternal generation were divided into two groups and given 0 (PND60a) and 4 mg/mL (PND60b) aluminum-lactate in their drinking water until PND 60. Significant neuronal injuries of hippocampus as well as a reduction in the m6A RNA modification and the expression of methylase were observed at PND 21 and PND 60a mice. The results indicated that Al-induced developmental neurotoxicity could persist into adulthood despite no sustained Al accumulation. m6A RNA modification had a crucial role in developmental neurotoxicity induced by Al. In addition, Al exposure during the embryonic to adult stages can cause more severe nerve damage and decline of m6A RNA modification. Collectively, these results suggest that the mechanism underlying Al-induced neurotoxicity appears to involve m6A RNA modification.


Subject(s)
Hippocampus , Neurons , RNA Methylation , Animals , Female , Mice , Pregnancy , Aluminum/toxicity , Hippocampus/drug effects , Hippocampus/growth & development , Hippocampus/metabolism , Hippocampus/pathology , Methyltransferases/genetics , Methyltransferases/metabolism , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/etiology , Prenatal Exposure Delayed Effects
18.
Food Chem Toxicol ; 191: 114851, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38986832

ABSTRACT

The issue of MeHg contamination is a significant concern due to its detrimental impact on the environment. This study aimed to thoroughly investigate the effects of MeHg on neurodevelopmental biomarkers, as there is a lack of systematic reviews in this area. We conducted a comprehensive search of three databases (PubMed, Scopus, and Web of Science) and found 522 records, which were then meticulously reviewed by two independent reviewers. A total of 66 studies were included, with biomarkers related to oxidative stress, neurotransmission, inflammation, epigenetics, and apoptosis being the most prominent. The results of both in vitro and in vivo models indicate that antioxidant enzymes and other oxidative stress-related markers are indeed, altered following MeHg exposure. Moreover, MeHg exposure causes significant disruptions to neurotransmitter levels, activities of neurotransmitter synthesis enzymes, receptor densities, and proteins involved in synaptic function. Proinflammatory biomarkers are consistently overexpressed in both MeHg-treated cells and the brains of exposed rats. Furthermore, studies on DNA methylation and biomarker activity suggest that MeHg exposure may lead to neurotoxicity and neurodevelopmental issues via perturbations to epigenetic markers and the apoptosis pathway.


Subject(s)
Biomarkers , Methylmercury Compounds , Oxidative Stress , Methylmercury Compounds/toxicity , Biomarkers/metabolism , Animals , Humans , Oxidative Stress/drug effects , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/etiology , Apoptosis/drug effects , Rats , Epigenesis, Genetic/drug effects , DNA Methylation/drug effects , Neurodevelopmental Disorders/chemically induced , Neurodevelopmental Disorders/metabolism
19.
Eur J Neurol ; 31(9): e16369, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38952074

ABSTRACT

BACKGROUND AND PURPOSE: A real-time biomarker in chemotherapy-induced peripheral neurotoxicity (CIPN) would be useful for clinical decision-making during treatment. Neurofilament light chain (NfL) can be detected in blood in the case of neuroaxonal damage. The aim of the study was to compare the levels of plasma NfL (pNfL) according to the type of chemotherapeutic agent and the severity of CIPN. METHODS: This single-center prospective observational longitudinal study included patients treated with paclitaxel (TX; n = 34), brentuximab vedotin (BV; n = 29), or oxaliplatin (PT; n = 19). All patients were assessed using the Total Neuropathy Score-clinical version and Common Terminology Criteria for Adverse Events before, during, and up to 6-12 months after the end of treatment. Nerve conduction studies (NCS) were performed before and after chemotherapy discontinuation. Consecutive plasma samples were analyzed for NfL levels using a Simoa® analyzer. Changes in pNfL were compared between groups and were eventually correlated with clinical and NCS data. Clinically relevant (CR) CIPN was considered to be grade ≥ 2. RESULTS: Eighty-two patients, mostly women (59.8%), were included. One third of the patients who received TX (29.4%), BV (31%), or PT (36.8%) developed CR-CIPN, respectively, without differences among them (p = 0.854). Although pNfL significantly increased during treatment and decreased throughout the recovery period in all three groups, patients receiving TX showed significantly greater and earlier changes in pNfL levels compared to the other agents (p < 0.001). CONCLUSIONS: A variable change in pNfL is observed depending on the type of agent and mechanism of neurotoxicity with comparable CIPN severity, strongly implying the need to identify different cutoff values for each agent.


Subject(s)
Antineoplastic Agents , Neurofilament Proteins , Neurotoxicity Syndromes , Peripheral Nervous System Diseases , Humans , Female , Male , Middle Aged , Neurofilament Proteins/blood , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/blood , Aged , Adult , Antineoplastic Agents/adverse effects , Longitudinal Studies , Neurotoxicity Syndromes/blood , Neurotoxicity Syndromes/etiology , Prospective Studies , Biomarkers/blood , Oxaliplatin/adverse effects , Paclitaxel/adverse effects
20.
Neurotoxicol Teratol ; 104: 107369, 2024.
Article in English | MEDLINE | ID: mdl-38964665

ABSTRACT

Pethoxamid, a member of the chloroacetamide herbicide family, is a recently approved chemical for pre- or post-emergence weed control; however, toxicity data for sublethal effects in aquatic organisms exposed to pethoxamid are non-existent in literature. To address this, we treated zebrafish embryos/larvae to pethoxamid over a 7-day period post-fertilization and evaluated several toxicological endpoints associated with oxidative stress and neurotoxicity. Continuous pethoxamid exposure did not affect survival nor hatch success in embryos/larvae for 7 days up to 1000 µg L-1. Exposure to pethoxamid did not affect embryonic ATP-linked respiration, but it did reduce non-mitochondrial respiration at the highest concentration tested. We also noted a significant increase in both apoptosis and levels of reactive oxygen species (ROS) in larvae zebrafish following exposure to pethoxamid. Increases in apoptosis and ROS, however, were not correlated with any altered gene expression pattern for apoptotic and oxidative damage response transcripts. To assess neurotoxicity potential, we measured behavior and several transcripts implicated in neural processes in the central nervous system. While locomotor activity of larval zebrafish was affected by pethoxamid exposure (hyperactivity was observed at concentrations below 1 µg L-1, and hypoactivity was noted at higher exposures to 10 and 100 µg L-1 pethoxamid), there were no effects on steady state mRNA abundance for neurotoxicity-related transcripts tested. This data contributes to knowledge regarding exposure risks for chloroacetamide-based herbicides and is the first study investigating sublethal toxicity for this newly registered herbicide.


Subject(s)
Apoptosis , Embryo, Nonmammalian , Herbicides , Larva , Oxidative Stress , Reactive Oxygen Species , Zebrafish , Animals , Zebrafish/embryology , Herbicides/toxicity , Embryo, Nonmammalian/drug effects , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Larva/drug effects , Oxidative Stress/drug effects , Acetamides/toxicity , Neurotoxicity Syndromes/etiology
SELECTION OF CITATIONS
SEARCH DETAIL