1
|
Xu SC, Zhong Y, Jiang HY, Tang J. Exposure to anti-seizure medication during pregnancy and the risk of autism and ADHD in offspring: a systematic review and meta-analysis. Front Neurol 2024; 15:1440145. [PMID: 39105059 PMCID: PMC11298387 DOI: 10.3389/fneur.2024.1440145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/10/2024] [Indexed: 08/07/2024] Open
Abstract
Background Evidence of an association between maternal use of anti-seizure medication (ASM) during pregnancy and the risk of autism spectrum disorder (ASD) or attention-deficit/hyperactivity disorder (ADHD) in children is conflicting. This systematic review and meta-analysis aimed to summarize the relationship between fetal exposure to ASM and the development of ASD or ADHD in offspring. Methods A comprehensive literature search was conducted in PubMed and other databases to identify relevant epidemiological studies published from inception until 1 March 2024. Results Seven cohort studies were included in the meta-analysis. The results showed that maternal exposure to ASMs during pregnancy was associated with an increased risk of ASD [odds ratio (OR): 2.1, 95% confidence interval (CI): 1.63-2.71; p < 0.001] in the general population. This association became weaker (ASD: OR: 1.38, 95% CI: 1.11-1.73; p = 0.004) when the reference group was mothers with a psychiatric disorder or epilepsy not treated during pregnancy. Furthermore, an increased risk of ADHD was observed when the study data adjusted for drug indications were pooled (OR: 1.43, 95% CI: 1.07-1.92; p = 0.015). In subgroup analyses based on individual ASM use, only exposure to valproate preconception was significantly associated with an increased risk of ASD or ADHD. Conclusion The significant association between maternal ASM use during pregnancy and ASD or ADHD in offspring may be partially explained by the drug indication or driven by valproate.
Collapse
Affiliation(s)
- Shan-Chun Xu
- Department of General Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Ying Zhong
- Department of Orthopaedics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Hai-Yin Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jun Tang
- Department of General Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
He C, Zhou H, Chen L, Liu Z. NEAT1 Promotes Valproic Acid-Induced Autism Spectrum Disorder by Recruiting YY1 to Regulate UBE3A Transcription. Mol Neurobiol 2024:10.1007/s12035-024-04309-y. [PMID: 38922486 DOI: 10.1007/s12035-024-04309-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024]
Abstract
Evidence suggests that long non-coding RNAs (lncRNAs) play a significant role in autism. Herein, we explored the functional role and possible molecular mechanisms of NEAT1 in valproic acid (VPA)-induced autism spectrum disorder (ASD). A VPA-induced ASD rat model was constructed, and a series of behavioral tests were performed to examine motor coordination and learning-memory abilities. qRT-PCR and western blot assays were used to evaluate target gene expression levels. Loss-and-gain-of-function assays were conducted to explore the functional role of NEAT1 in ASD development. Furthermore, a combination of mechanistic experiments and bioinformatic tools was used to assess the relationship and regulatory role of the NEAT1-YY1-UBE3A axis in ASD cellular processes. Results showed that VPA exposure induced autism-like developmental delays and behavioral abnormalities in the VPA-induced ASD rat model. We found that NEAT1 was elevated in rat hippocampal tissues after VPA exposure. NEAT1 promoted VPA-induced autism-like behaviors and mitigated apoptosis, oxidative stress, and inflammation in VPA-induced ASD rats. Notably, NEAT1 knockdown improved autism-related behaviors and ameliorated hippocampal neuronal damage. Mechanistically, it was observed that NEAT1 recruited the transcription factor YY1 to regulate UBE3A expression. Additionally, in vitro experiments further confirmed that NEAT1 knockdown mitigated hippocampal neuronal damage, oxidative stress, and inflammation through the YY1/UBE3A axis. In conclusion, our study demonstrates that NEAT1 is highly expressed in ASD, and its inhibition prominently suppresses hippocampal neuronal injury and oxidative stress through the YY1/UBE3A axis, thereby alleviating ASD development. This provides a new direction for ASD-targeted therapy.
Collapse
Affiliation(s)
- Chuping He
- Department of Children's Health, Chenzhou First People's Hospital, No. 6, Feihong Road, Suxian District, Chenzhou, 423000, Hunan, China
| | - Huimei Zhou
- Department of Children's Health, Chenzhou First People's Hospital, No. 6, Feihong Road, Suxian District, Chenzhou, 423000, Hunan, China.
| | - Lei Chen
- Department of Children's Health, Chenzhou First People's Hospital, No. 6, Feihong Road, Suxian District, Chenzhou, 423000, Hunan, China
| | - Zeying Liu
- Department of Children's Health, Chenzhou First People's Hospital, No. 6, Feihong Road, Suxian District, Chenzhou, 423000, Hunan, China
| |
Collapse
|
3
|
Chen A, Wang M, Xu C, Zhao Y, Xian P, Li Y, Zheng W, Yi X, Wu S, Wang Y. Glycolysis mediates neuron specific histone acetylation in valproic acid-induced human excitatory neuron differentiation. Front Mol Neurosci 2023; 16:1151162. [PMID: 37089691 PMCID: PMC10118002 DOI: 10.3389/fnmol.2023.1151162] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/20/2023] [Indexed: 04/08/2023] Open
Abstract
Pregnancy exposure of valproic acid (VPA) is widely adopted as a model of environmental factor induced autism spectrum disorder (ASD). Increase of excitatory/inhibitory synaptic transmission ratio has been proposed as the mechanism of VPA induced ASD. How this happened, particularly at the level of excitatory neuron differentiation in human neural progenitor cells (NPCs) remains largely unclear. Here, we report that VPA exposure remarkably inhibited human NPC proliferation and induced excitatory neuronal differentiation without affecting inhibitory neurons. Following VPA treatment, mitochondrial dysfunction was observed before neuronal differentiation, as showed by ultrastructural changes, respiratory complex activity, mitochondrial membrane potential and oxidation levels. Meanwhile, extracellular acidification assay revealed an elevation of glycolysis by VPA stimulation. Interestingly, inhibiting glycolysis by 2-deoxy-d-glucose-6-phosphate (2-DG) efficiently blocked the excitatory neuronal differentiation of human NPCs induced by VPA. Furthermore, 2-DG treatment significantly compromised the VPA-induced expression of H3ac and H3K9ac, and the VPA-induced binding of H3K9ac on the promoter of Ngn2 and Mash1, two key transcription factors of excitatory neuron fate determination. These data, for the first time, demonstrated that VPA biased excitatory neuron differentiation by glycolysis-mediated histone acetylation of neuron specific transcription factors.
Collapse
Affiliation(s)
- Andi Chen
- Department of Neurobiology, School of Basic Medicine, Institute of Neurosciences, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Mengmeng Wang
- Department of Neurobiology, School of Basic Medicine, Institute of Neurosciences, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Chao Xu
- Department of Neurobiology, School of Basic Medicine, Institute of Neurosciences, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Youyi Zhao
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research, Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Panpan Xian
- Department of Neurobiology, School of Basic Medicine, Institute of Neurosciences, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yuqian Li
- Department of Neurobiology, School of Basic Medicine, Institute of Neurosciences, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Weian Zheng
- Department of Neurobiology, School of Basic Medicine, Institute of Neurosciences, Fourth Military Medical University, Xi’an, Shaanxi, China
- School of Life Sciences and Research Center for Natural Peptide Drugs, Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, Yan’an University, Yan’an, China
| | - Xuyang Yi
- Department of Neurobiology, School of Basic Medicine, Institute of Neurosciences, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Institute of Neurosciences, Fourth Military Medical University, Xi’an, Shaanxi, China
- Shengxi Wu,
| | - Yazhou Wang
- Department of Neurobiology, School of Basic Medicine, Institute of Neurosciences, Fourth Military Medical University, Xi’an, Shaanxi, China
- *Correspondence: Yazhou Wang,
| |
Collapse
|
4
|
Shnayder NA, Grechkina VV, Khasanova AK, Bochanova EN, Dontceva EA, Petrova MM, Asadullin AR, Shipulin GA, Altynbekov KS, Al-Zamil M, Nasyrova RF. Therapeutic and Toxic Effects of Valproic Acid Metabolites. Metabolites 2023; 13:metabo13010134. [PMID: 36677060 PMCID: PMC9862929 DOI: 10.3390/metabo13010134] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Valproic acid (VPA) and its salts are psychotropic drugs that are widely used in neurological diseases (epilepsy, neuropathic pain, migraine, etc.) and psychiatric disorders (schizophrenia, bipolar affective disorder, addiction diseases, etc.). In addition, the indications for the appointment of valproate have been expanding in recent years in connection with the study of new mechanisms of action of therapeutic and toxic metabolites of VPA in the human body. Thus, VPA is considered a component of disease-modifying therapy for multiple tumors, neurodegenerative diseases (Huntington's disease, Parkinson's disease, Duchenne progressive dystrophy, etc.), and human immunodeficiency syndrome. The metabolism of VPA is complex and continues to be studied. Known pathways of VPA metabolism include: β-oxidation in the tricarboxylic acid cycle (acetylation); oxidation with the participation of cytochrome P-450 isoenzymes (P-oxidation); and glucuronidation. The complex metabolism of VPA explains the diversity of its active and inactive metabolites, which have therapeutic, neutral, or toxic effects. It is known that some active metabolites of VPA may have a stronger clinical effect than VPA itself. These reasons explain the relevance of this narrative review, which summarizes the results of studies of blood (serum, plasma) and urinary metabolites of VPA from the standpoint of the pharmacogenomics and pharmacometabolomics. In addition, a new personalized approach to assessing the cumulative risk of developing VPA-induced adverse reactions is presented and ways for their correction are proposed depending on the patient's pharmacogenetic profile and the level of therapeutic and toxic VPA metabolites in the human body fluids (blood, urine).
Collapse
Affiliation(s)
- Natalia A. Shnayder
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
- Correspondence: (N.A.S.); (R.F.N.); Tel.: +7-(812)-620-0222 (N.A.S. & R.F.N.)
| | - Violetta V. Grechkina
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia
| | - Aiperi K. Khasanova
- Department of Psychiatry, Russian Medical Academy for Continual Professional Education, 125993 Moscow, Russia
| | - Elena N. Bochanova
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Evgenia A. Dontceva
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Marina M. Petrova
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Azat R. Asadullin
- Department of Psychiatry and Addiction, Bashkir State Medical University, 45000 Ufa, Russia
| | - German A. Shipulin
- Centre for Strategic Planning and Management of Biomedical Health Risks, 119121 Moscow, Russia
| | - Kuanysh S. Altynbekov
- Republican Scientific and Practical Center of Mental Health, Almaty 050022, Kazakhstan
- Department of Psychiatry and Narcology, S.D. Asfendiarov Kazakh National Medical University, Almaty 050022, Kazakhstan
| | - Mustafa Al-Zamil
- Department of Physiotherapy, Faculty of Continuing Medical Education, Peoples’ Friendship University of Russia, 11798 Moscow, Russia
| | - Regina F. Nasyrova
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia
- Correspondence: (N.A.S.); (R.F.N.); Tel.: +7-(812)-620-0222 (N.A.S. & R.F.N.)
| |
Collapse
|
5
|
Li Y, Gao S, Meng Y. Integrated analysis of endoplasmic reticulum stress regulators' expression identifies distinct subtypes of autism spectrum disorder. Front Psychiatry 2023; 14:1136154. [PMID: 37139330 PMCID: PMC10149679 DOI: 10.3389/fpsyt.2023.1136154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 03/24/2023] [Indexed: 05/05/2023] Open
Abstract
Endoplasmic reticulum (ER) stress has been demonstrated to play important roles in a variety of human diseases. However, their relevance to autism spectrum disorder (ASD) remains largely unknown. Herein, we aimed to investigate the expression patterns and potential roles of the ER stress regulators in ASD. The ASD expression profiles GSE111176 and GSE77103 were compiled from the Gene Expression Omnibus (GEO) database. ER stress score determined by the single sample gene set enrichment analysis (ssGSEA) was significantly higher in ASD patients. Differential analysis revealed that there were 37 ER stress regulators dysregulated in ASD. Based on their expression profile, the random forest and artificial neuron network techniques were applied to build a classifier that can effectively distinguish ASD from control samples among independent datasets. Weighted gene co-expression network analysis (WGCNA) screened out the turquoise module with 774 genes was closely related to the ER stress score. Through the overlapping results of the turquoise module and differential expression ER stress genes, hub regulators were gathered. The TF/miRNA-hub gene interaction networks were created. Furthermore, the consensus clustering algorithm was performed to cluster the ASD patients, and there were two ASD subclusters. Each subcluster has unique expression profiles, biological functions, and immunological characteristics. In ASD subcluster 1, the FAS pathway was more enriched, while subcluster 2 had a higher level of plasma cell infiltration as well as the BCR signaling pathway and interleukin receptor reaction reactivity. Finally, the Connectivity map (CMap) database was used to find prospective compounds that target various ASD subclusters. A total of 136 compounds were significantly enriched. In addition to some specific drugs which can effectively reverse the differential gene expression of each subcluster, we found that the PKC inhibitor BRD-K09991945 that targets Glycogen synthase kinase 3β (GSK3B) might have a therapeutic effect on both ASD subtypes that worth of the experimental validation. Our finding proved that ER stress plays a crucial role in the diversity and complexity of ASD, which may inform both mechanistic and therapeutic assessments of the disorder.
Collapse
|
6
|
Unnisa A, Greig NH, Kamal MA. Modelling the Interplay Between Neuron-Glia Cell Dysfunction and Glial Therapy in Autism Spectrum Disorder. Curr Neuropharmacol 2023; 21:547-559. [PMID: 36545725 PMCID: PMC10207919 DOI: 10.2174/1570159x21666221221142743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/27/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
Autism spectrum disorder (ASD) is a complicated, interpersonally defined, static condition of the underdeveloped brain. Although the aetiology of autism remains unclear, disturbance of neuronglia interactions has lately been proposed as a significant event in the pathophysiology of ASD. In recent years, the contribution of glial cells to autism has been overlooked. In addition to neurons, glial cells play an essential role in mental activities, and a new strategy that emphasises neuron-glia interactions should be applied. Disturbance of neuron-glia connections has lately been proposed as a significant event in the pathophysiology of ASD because aberrant neuronal network formation and dysfunctional neurotransmission are fundamental to the pathology of the condition. In ASD, neuron and glial cell number changes cause brain circuits to malfunction and impact behaviour. A study revealed that reactive glial cells result in the loss of synaptic functioning and induce autism under inflammatory conditions. Recent discoveries also suggest that dysfunction or changes in the ability of microglia to carry out physiological and defensive functions (such as failure in synaptic elimination or aberrant microglial activation) may be crucial for developing brain diseases, especially autism. The cerebellum, white matter, and cortical regions of autistic patients showed significant microglial activation. Reactive glial cells result in the loss of synaptic functioning and induce autism under inflammatory conditions. Replacement of defective glial cells (Cell-replacement treatment), glial progenitor cell-based therapy, and medication therapy (inhibition of microglia activation) are all utilised to treat glial dysfunction. This review discusses the role of glial cells in ASD and the various potential approaches to treating glial cell dysfunction.
Collapse
Affiliation(s)
- Aziz Unnisa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail, KSA;
| | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, MD 21224, USA
| | - Mohammad Amjad Kamal
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, China
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
- Enzymoics, Novel Global Community Educational Foundation, Peterlee place, Hebersham, NSW 2770, Australia
| |
Collapse
|
7
|
Mehra S, Ul Ahsan A, Seth E, Chopra M. Critical Evaluation of Valproic Acid-Induced Rodent Models of Autism: Current and Future Perspectives. J Mol Neurosci 2022; 72:1259-1273. [DOI: 10.1007/s12031-022-02033-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 05/21/2022] [Indexed: 11/29/2022]
|
8
|
GENDER DIFFERENCES IN EFFECTS OF PRENATAL AND POSTNATAL EXPOSURE TO ELECTROMAGNETIC FIELD AND PRENATAL ZINC ON BEHAVIOUR AND SYNAPTIC PROTEINS IN RATS. J Chem Neuroanat 2022; 122:102092. [DOI: 10.1016/j.jchemneu.2022.102092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 11/21/2022]
|
9
|
Traetta ME, Uccelli NA, Zárate SC, Gómez Cuautle D, Ramos AJ, Reinés A. Long-Lasting Changes in Glial Cells Isolated From Rats Subjected to the Valproic Acid Model of Autism Spectrum Disorder. Front Pharmacol 2021; 12:707859. [PMID: 34421599 PMCID: PMC8374432 DOI: 10.3389/fphar.2021.707859] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/29/2021] [Indexed: 01/01/2023] Open
Abstract
Synaptic alterations concomitant with neuroinflammation have been described in patients and experimental models of autism spectrum disorder (ASD). However, the role of microglia and astroglia in relation to synaptic changes is poorly understood. Male Wistar rats prenatally exposed to valproic acid (VPA, 450 mg/kg, i.p.) or saline (control) at embryonic day 10.5 were used to study synapses, microglia, and astroglia in the prefrontal cortex (PFC) at postnatal days 3 and 35 (PND3 and PND35). Primary cultures of cortical neurons, microglia, and astroglia isolated from control and VPA animals were used to study each cell type individually, neuron-microglia and microglia-astroglia crosstalk. In the PFC of VPA rats, synaptic changes characterized by an increase in the number of excitatory synapses were evidenced at PND3 and persisted until PND35. At PND3, microglia and astroglia from VPA animals were morphologically similar to those of age-matched controls, whereas at PND35, reactive microgliosis and astrogliosis were observed in the PFC of VPA animals. Cortical neurons isolated from VPA rats mimicked in vitro the synaptic pattern seen in vivo. Cortical microglia and astroglia isolated from VPA animals exhibited reactive morphology, increased pro-inflammatory cytokines, and a compromised miRNA processing machinery. Microglia from VPA animals also showed resistance to a phagocytic challenge. In the presence of neurons from VPA animals, microglia isolated from VPA rats revealed a non-reactive morphology and promoted neurite outgrowth, while microglia from control animals displayed a reactive profile and promoted dendritic retraction. In microglia-astroglia co-cultures, microglia from VPA animals displayed a reactive profile and exacerbated astrocyte reactivity. Our study indicates that cortical microglia from VPA animals are insensitive or adapted to neuronal cues expressed by neurons from VPA animals. Further, long-term in vivo microgliosis could be the result of altered microglia-astroglia crosstalk in VPA animals. Thus, our study highlights cortical microglia-astroglia communication as a new mechanism implicated in neuroinflammation in ASD; consequently, we propose that this crosstalk is a potential target for interventions in this disorder.
Collapse
Affiliation(s)
- Marianela Evelyn Traetta
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Cátedra de Farmacología, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nonthué Alejandra Uccelli
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sandra Cristina Zárate
- Facultad de Medicina, Departamento de Histología, Embriología, Biología Celular y Genética, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Investigaciones Biomédicas (INBIOMED), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Dante Gómez Cuautle
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alberto Javier Ramos
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina.,Facultad de Medicina, Departamento de Histología, Embriología, Biología Celular y Genética, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Analía Reinés
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Cátedra de Farmacología, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|