Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where María Elisa Alonso is active.

Publication


Featured researches published by María Elisa Alonso.


Nature Genetics | 2004

Mutations in EFHC1 cause juvenile myoclonic epilepsy

Toshimitsu Suzuki; Antonio V. Delgado-Escueta; Kripamoy Aguan; María Elisa Alonso; Jun Shi; Yuji Hara; M Nishida; Tomohiro Numata; Marco T. Medina; Tamaki Takeuchi; Ryoji Morita; Dongsheng Bai; Subramaniam Ganesh; Yoshihisa Sugimoto; Johji Inazawa; Julia N. Bailey; Adriana Ochoa; Aurelio Jara-Prado; Astrid Rasmussen; Jaime Ramos-Peek; Sergio Cordova; Francisco Rubio-Donnadieu; Yushi Inoue; Makiko Osawa; Sunao Kaneko; Hirokazu Oguni; Yasuo Mori; Kazuhiro Yamakawa

Juvenile myoclonic epilepsy (JME) is the most frequent cause of hereditary grand mal seizures. We previously mapped and narrowed a region associated with JME on chromosome 6p12–p11 (EJM1). Here, we describe a new gene in this region, EFHC1, which encodes a protein with an EF-hand motif. Mutation analyses identified five missense mutations in EFHC1 that cosegregated with epilepsy or EEG polyspike wave in affected members of six unrelated families with JME and did not occur in 382 control individuals. Overexpression of EFHC1 in mouse hippocampal primary culture neurons induced apoptosis that was significantly lowered by the mutations. Apoptosis was specifically suppressed by SNX-482, an antagonist of R-type voltage-dependent Ca2+ channel (Cav2.3). EFHC1 and Cav2.3 immunomaterials overlapped in mouse brain, and EFHC1 coimmunoprecipitated with the Cav2.3 C terminus. In patch-clamp analysis, EFHC1 specifically increased R-type Ca2+ currents that were reversed by the mutations associated with JME.


American Journal of Human Genetics | 2008

Hyperglycosylation and Reduced GABA Currents of Mutated GABRB3 Polypeptide in Remitting Childhood Absence Epilepsy

Miyabi Tanaka; Richard W. Olsen; Marco T. Medina; Emily Schwartz; María Elisa Alonso; Reyna M. Durón; Ramon Castro-Ortega; Iris E. Martínez-Juárez; Ignacio Pascual-Castroviejo; Jesús Machado-Salas; Rene Silva; Julia N. Bailey; Dongsheng Bai; Adriana Ochoa; Aurelio Jara-Prado; Gregorio Pineda; Robert L. Macdonald; Antonio V. Delgado-Escueta

Childhood absence epilepsy (CAE) accounts for 10% to 12% of epilepsy in children under 16 years of age. We screened for mutations in the GABA(A) receptor (GABAR) beta 3 subunit gene (GABRB3) in 48 probands and families with remitting CAE. We found that four out of 48 families (8%) had mutations in GABRB3. One heterozygous missense mutation (P11S) in exon 1a segregated with four CAE-affected persons in one multiplex, two-generation Mexican family. P11S was also found in a singleton from Mexico. Another heterozygous missense mutation (S15F) was present in a singleton from Honduras. An exon 2 heterozygous missense mutation (G32R) was present in two CAE-affected persons and two persons affected with EEG-recorded spike and/or sharp wave in a two-generation Honduran family. All mutations were absent in 630 controls. We studied functions and possible pathogenicity by expressing mutations in HeLa cells with the use of Western blots and an in vitro translation and translocation system. Expression levels did not differ from those of controls, but all mutations showed hyperglycosylation in the in vitro translation and translocation system with canine microsomes. Functional analysis of human GABA(A) receptors (alpha 1 beta 3-v2 gamma 2S, alpha 1 beta 3-v2[P11S]gamma 2S, alpha 1 beta 3-v2[S15F]gamma 2S, and alpha 1 beta 3-v2[G32R]gamma 2S) transiently expressed in HEK293T cells with the use of rapid agonist application showed that each amino acid transversion in the beta 3-v2 subunit (P11S, S15F, and G32R) reduced GABA-evoked current density from whole cells. Mutated beta 3 subunit protein could thus cause absence seizures through a gain in glycosylation of mutated exon 1a and exon 2, affecting maturation and trafficking of GABAR from endoplasmic reticulum to cell surface and resulting in reduced GABA-evoked currents.


American Journal of Medical Genetics | 1996

Juvenile myoclonic epilepsy in chromosome 6p12‐p11: Locus heterogeneity and recombinations

A. W. Liu; Antonio V. Delgado-Escueta; M. N. Gee; J. M. Serratosa; Q. W. Zhang; María Elisa Alonso; Marco T. Medina; Sergio Cordova; H. Z. Zhao; J. M. Spellman; F. Rubio Donnadieu; J. Ramos Peek; Lucy J. Treiman; R. S. Sparkes

We recently analyzed under homogeneity a large pedigree from Belize with classic juvenile myoclonic epilepsy (JME). After a genome wide search with 146 microsatellites, we obtained significant linkage between chromosome 6p markers, D6S257 and D6S272, and both convulsive and EEG traits of JME. Recombinations in two affected members defined a 40 cM JME region flanked by D6S313 and D6S258. In the present communication, we explored if the same chromosome 6p11 microsatellites also have a role in JME mixed with pyknoleptic absences. We allowed for heterogeneity during linkage analyses. We tested for heterogeneity by the admixture test and looked for more recombinations. D6S272, D6S466, D6S294, and D6S257 were significantly linked (Zmax > 3.5) to the clinical and EEG traits of 22 families, assuming autosomal dominant inheritance with 70% penetrance. Pairwise Zmax were 4.230 for D6S294 (theta m = f at 0.133) and 4.442 for D6S466 (theta m = f at 0.111). Admixture test (H2 vs. H1) was significant (P = 0.0234 for D6S294 and 0.0128 for D6S272) supporting the hypotheses of linkage with heterogeneity. Estimated proportion of linked families, alpha, was 0.50 (95% confidence interval 0.05-0.99) for D6S294 and D6S272. Multipoint analyses and recombinations in three new families narrowed the JME locus to a 7 cM interval flanked by D6S272 and D6S257.


Neurology | 2008

Novel mutations in Myoclonin1/EFHC1 in sporadic and familial juvenile myoclonic epilepsy

Mayerlim Medina; Toshimitsu Suzuki; María Elisa Alonso; Reyna M. Durón; Iris E. Martínez-Juárez; Julia N. Bailey; Dongsheng Bai; Yushi Inoue; I. Yoshimura; Sunao Kaneko; M. C. Montoya; Adriana Ochoa; A. Jara Prado; Miyabi Tanaka; Jesús Machado-Salas; S. Fujimoto; M. Ito; S. Hamano; K. Sugita; Y. Ueda; Makiko Osawa; Hirokazu Oguni; Francisco Rubio-Donnadieu; Kazuhiro Yamakawa; Antonio V. Delgado-Escueta

Background: Juvenile myoclonic epilepsy (JME) accounts for 3 to 12% of all epilepsies. In 2004, the GENESS Consortium demonstrated four missense mutations in Myoclonin1/EFHC1 of chromosome 6p12.1 segregating in 20% of Hispanic families with JME. Objective: To examine what percentage of consecutive JME clinic cases have mutations in Myoclonin1/EFHC1. Methods: We screened 44 consecutive patients from Mexico and Honduras and 67 patients from Japan using heteroduplex analysis and direct sequencing. Results: We found five novel mutations in transcripts A and B of Myoclonin1/EFHC1. Two novel heterozygous missense mutations (c.755C>A and c.1523C>G) in transcript A occurred in both a singleton from Mexico and another singleton from Japan. A deletion/frameshift (C.789del.AV264fsx280) in transcript B was present in a mother and daughter from Mexico. A nonsense mutation (c.829C>T) in transcript B segregated in four clinically and seven epileptiform-EEG affected members of a large Honduran family. The same nonsense mutation (c.829C>T) occurred as a de novo mutation in a sporadic case. Finally, we found a three-base deletion (−364○%–362del.GAT) in the promoter region in a family from Japan. Conclusion: Nine percent of consecutive juvenile myoclonic epilepsy cases from Mexico and Honduras clinics and 3% of clinic patients from Japan carry mutations in Myoclonin1/EFCH1. These results represent the highest number and percentage of mutations found for a juvenile myoclonic epilepsy causing gene of any population group. GLOSSARY: CAE = childhood absence epilepsy; FS = febrile seizures in infancy/childhood; GM = grand mal tonic clonic seizure; JME = Juvenile myoclonic epilepsy; PSW = 3–6 Hz polyspike and slow wave complexes; SW = single spike and slow wave complex.


Epilepsia | 2005

Seizures of Idiopathic Generalized Epilepsies

Reyna M. Durón; Marco T. Medina; Iris E. Martínez-Juárez; Julia N. Bailey; Perez-Gosiengfiao Kt; Ricardo Ramos-Ramírez; Minerva López-Ruiz; María Elisa Alonso; Ramón H. Castro Ortega; Ignacio Pascual‐Castroviejo; Jesús Machado-Salas; Lizardo Mija; Antonio V. Delgado-Escueta

Summary:  Idiopathic generalized epilepsies (IGEs) comprise at least 40% of epilepsies in the United States, 20% in Mexico, and 8% in Central America. Here, we review seizure phenotypes across IGE syndromes, their response to treatment and advances in molecular genetics that influence nosology. Our review included the Medline database from 1945 to 2005 and our prospectively collected Genetic Epilepsy Studies (GENESS) Consortium database. Generalized seizures occur with different and similar semiologies, frequencies, and patterns, ages at onset, and outcomes in different IGEs, suggesting common neuroanatomical pathways for seizure phenotypes. However, the same seizure phenotypes respond differently to the same treatments in different IGEs, suggesting different molecular defects across syndromes. De novo mutations in SCN1A in sporadic Dravet syndrome and germline mutations in SCN1A, SCN1B, and SCN2A in generalized epilepsies with febrile seizures plus have unraveled the heterogenous myoclonic epilepsies of infancy and early childhood. Mutations in GABRA1, GABRG2, and GABRB3 are associated with absence seizures, while mutations in CLCN2 and myoclonin/EFHC1 substantiate juvenile myoclonic epilepsy as a clinical entity. Refined understanding of seizure phenotypes, their semiology, frequencies, and patterns together with the identification of molecular lesions in IGEs continue to accelerate the development of molecular epileptology.


European Journal of Clinical Pharmacology | 2005

CYP2D6 genotype and phenotype determination in a Mexican Mestizo population

Marisol López; Jorge Guerrero; Helgi Jung–Cook; María Elisa Alonso

ObjectiveAlthough CYP2D6 genetic polymorphism plays an important role in interindividual and interethnic variability in drug response, very few pharmacogenetic data are available from Hispanic populations, including Mexicans. For this purpose, this study was undertaken to determine CYP2D6 genotype and phenotype in a healthy Mexican Mestizo population.MethodsGenotyping of five CYP2D6 mutant alleles by PCR–RFLP, and CYP2D6*5 and duplicated CYP2D6 alleles by long-PCR was performed in two hundred and forty three Mexican Mestizos. Of these, one hundred subjects were also phenotyped using dextromethorphan as the probe drug.ResultsThe frequency of CYP2D6*2, *3, *4, *5, *10, *17 was 19.34%, 1.44%, 11.21%, 2.67%, 12.45%, and 1.65%, respectively, while duplicated CYP2D6 alleles were found in 12.76% of the 243 genotyped subjects. Among the 100 phenotyped subjects, we identified ten (10%, 95% confidence interval of 4.12–15.9) individuals as poor metabolizers by using the published antimode for Caucasians. The mean log10 dextromethorphan/dextrorphan ratio of the total sample was −2.05. The mean (SD) of the log10 MR in the CYP2D6 subgroups was UM=−2.6 (0.86); EM=−2.09 (0.98); IM=−1.71 (1.06); and PM=0.42 (0.625). These data show a trend toward a smaller mean log MR (higher enzyme activity) as the number of active alleles increases.ConclusionsThe PM frequency of CYP2D6 in the population studied was 10%, which is very similar to Spanish Caucasians. The observed frequency of the CYP2D6 alleles tested was unique for the Mexican Mestizo sample analyzed, and in accordance to the Caucasian, Asian and African admixture in this population.


Movement Disorders | 2009

Clinical and genetic characteristics of Mexican Huntington's disease patients.

María Elisa Alonso; Adriana Ochoa; Marie-Catherine Boll; Ana Luisa Sosa; Petra Yescas; Marisol López; Rosario Macias; Itziar Familiar; Astrid Rasmussen

We report the characteristics of 691 Mexican patients with Huntingtons disease (HD). These patients, representing 401 families, constitute the largest series of Mexican HD cases as yet described in the literature. We found the clinical characteristics of these patients to be similar to those of other populations, but we observed a higher frequency of infantile cases, a shorter disease duration and a lower suicide rate. In 626 cases, for which molecular analyses were available, CAG‐trinucleotide expansion size ranged from 37–106 repeats. The large number of CAG repeats (19.04 ± 3.02) in normal alleles and the presence of new mutations suggest that the overall prevalence of HD in the Mexican population could be expected to be within range of, or higher than, that reported for Europeans.


Neurogenetics | 2006

Founder effect for the Ala431Glu mutation of the presenilin 1 gene causing early-onset Alzheimer's disease in Mexican families.

Petra Yescas; Adriana Huertas-Vazquez; María Teresa Villarreal-Molina; Astrid Rasmussen; María Teresa Tusié-Luna; Marisol López; Samuel Canizales-Quinteros; María Elisa Alonso

The etiology of Alzheimer’s disease (AD) is complex. To date, molecular genetic studies in several families affected with AD have identified three genes associated with highly penetrant early-onset AD: Presenilin 1 (PSEN1), Presenilin 2 (PSEN2) and β-amyloid precursor protein (APP); and one gene (apolipoprotein E) associated with late-onset AD. Molecular analysis of the PSEN1 gene was performed by direct sequencing of genomic DNA. The possible founder effect was investigated analyzing two highly polymorphic microsatellite markers flanking the PSEN1 gene. Twelve unrelated Mexican families with early-onset AD were analyzed. The Ala431Glu mutation in exon 12 of PSEN1 was found in nine (75%) of these families, which segregated showing autosomal dominant inheritance. Because all families bearing the mutation are from the State of Jalisco (located in Western Mexico), a founder effect was hypothesized. Microsatellite haplotype analysis suggested a common ancestor in these nine kindreds. In conclusion, the Ala431Glu mutation is a prevalent cause of early-onset familial Alzheimer’s disease in families from the State of Jalisco, Mexico. Genetic evidence supports that it is a founder mutation descending from a single common ancestor. These findings have important implications for prompt diagnosis and genetic counseling for Mexican patients with familial AD from Jalisco.


Drug metabolism and drug interactions | 2011

Pharmacogenetics of the antiepileptic drugs phenytoin and lamotrigine.

Marisol López; Pedro Dorado; Nancy Monroy; María Elisa Alonso; Helgi Jung-Cook; Esther Machín; Eva Peñas-Lledó; Adrián LLerena

Abstract Patients treated with antiepileptic drugs can exhibit large interindividual variability in clinical efficacy or adverse effects. This could be partially due to genetic variants in genes coding for proteins that function as drug metabolizing enzymes, drug transporters or drug targets. The purpose of this article is to provide an overview of the current knowledge on the pharmacogenetics of two commonly prescribed antiepileptic drugs with similar mechanisms of action; phenytoin (PHT) and lamotrigine (LTG). These two drugs have been selected in order to model the pharmacogenetics of Phase I and Phase II metabolism for PHT and LTG, respectively. In light of the present evidence, patients treated with PHT could benefit from CYP2C9 and CYP2C19 genotyping/phenotyping. For those under treatment with LTG, UGT1A4 and UGT2B7 genotyping might be of clinical use and could contribute to the interindividual variability in LTG concentration to dose ratio in epileptic patients.


Epilepsia | 2003

Recent Developments in the Quest for Myoclonic Epilepsy Genes

Antonio V. Delgado-Escueta; Katerina Perez-Gosiengfiao; Dongsheng Bai; Julia N. Bailey; Marco T. Medina; Ryoji Morita; Toshimitsu Suzuki; Subramaniam Ganesh; Toshihisa Sugimoto; Kazuhiro Yamakawa; Adriana Ochoa; Aurelio Jara-Prado; Astrid Rasmussen; Jaime Ramos-Peek; Sergio Cordova; Francisco Rubio-Donnadieu; María Elisa Alonso

Summary:  Understanding the latest advances in the molecular genetics of the epilepsies is important, as it provides a basis for comprehending the new practice of epileptology. Epilepsies have traditionally been classified and subtyped on the basis of clinical and neurophysiologic concepts. However, the complexity and variability of phenotypes and overlapping clinical features limit the resolution of phenotype‐based classification and confound epilepsy nosology. Identification of tightly linked epilepsy DNA markers and discovery of epilepsy‐causing mutations provide a basis for refining the classification of epilepsies. Recent discoveries regarding the genetics surrounding certain epilepsy types (including Laforas progressive myoclonic epilepsy, the severe myoclonic epilepsy of infancy of Dravet, and idiopathic generalized epilepsies) may be the beginning of a better understanding of how rare Mendelian epilepsy genes and their genetic architecture can explain some complexities of the common epilepsies.

Collaboration


Dive into the María Elisa Alonso's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marco T. Medina

Universidad Nacional Autónoma de Honduras

View shared research outputs
Top Co-Authors

Avatar

Adriana Ochoa

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Astrid Rasmussen

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Kazuhiro Yamakawa

RIKEN Brain Science Institute

View shared research outputs
Top Co-Authors

Avatar

Dongsheng Bai

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge