Perfil de conectividad cerebral y el rendimiento cognitivo en consulta neurológica

El abordaje diagnóstico de pacientes con enfermedades que comprometen la cognición es un reto para los sistemas de salud. El electroencefalograma es considerado una técnica útil para evaluar el funcionamiento cerebral, de bajo costo y accesible a la población general. El presente estudio indica que...

Full description

Autores:
Camargo Camargo, Loida
Tipo de recurso:
Doctoral thesis
Fecha de publicación:
2023
Institución:
Corporación Universidad de la Costa
Repositorio:
REDICUC - Repositorio CUC
Idioma:
spa
OAI Identifier:
oai:repositorio.cuc.edu.co:11323/10766
Acceso en línea:
https://hdl.handle.net/11323/10766
https://repositorio.cuc.edu.co/
Palabra clave:
Demencia tipo alzheimer (DA)
Deterioro cognitivo leve (DCL)
Electroencefalograma (EEG)
Espectro de potencia
Conectividad funcional
Fototest
MoCA
Alzheimer's dementia (AD)
Mild cognitive impairment (MCI)
Electroencephalogram (EEG)
Power spectra
Functional connectivity
Rights
closedAccess
License
Atribución-NoComercial-CompartirIgual 4.0 Internacional (CC BY-NC-SA 4.0)
id RCUC2_e2cbc0f3cdacffad8bb50eff5abe1981
oai_identifier_str oai:repositorio.cuc.edu.co:11323/10766
network_acronym_str RCUC2
network_name_str REDICUC - Repositorio CUC
repository_id_str
dc.title.spa.fl_str_mv Perfil de conectividad cerebral y el rendimiento cognitivo en consulta neurológica
title Perfil de conectividad cerebral y el rendimiento cognitivo en consulta neurológica
spellingShingle Perfil de conectividad cerebral y el rendimiento cognitivo en consulta neurológica
Demencia tipo alzheimer (DA)
Deterioro cognitivo leve (DCL)
Electroencefalograma (EEG)
Espectro de potencia
Conectividad funcional
Fototest
MoCA
Alzheimer's dementia (AD)
Mild cognitive impairment (MCI)
Electroencephalogram (EEG)
Power spectra
Functional connectivity
title_short Perfil de conectividad cerebral y el rendimiento cognitivo en consulta neurológica
title_full Perfil de conectividad cerebral y el rendimiento cognitivo en consulta neurológica
title_fullStr Perfil de conectividad cerebral y el rendimiento cognitivo en consulta neurológica
title_full_unstemmed Perfil de conectividad cerebral y el rendimiento cognitivo en consulta neurológica
title_sort Perfil de conectividad cerebral y el rendimiento cognitivo en consulta neurológica
dc.creator.fl_str_mv Camargo Camargo, Loida
dc.contributor.advisor.none.fl_str_mv Allegri, Ricardo Francisco
Barceló, Ernesto
Cuesta, Pablo
dc.contributor.author.none.fl_str_mv Camargo Camargo, Loida
dc.contributor.jury.none.fl_str_mv Cochen, Silvia
Thomson, Alfredo
Fabian Román, Nestor
dc.subject.proposal.spa.fl_str_mv Demencia tipo alzheimer (DA)
Deterioro cognitivo leve (DCL)
Electroencefalograma (EEG)
Espectro de potencia
Conectividad funcional
Fototest
MoCA
topic Demencia tipo alzheimer (DA)
Deterioro cognitivo leve (DCL)
Electroencefalograma (EEG)
Espectro de potencia
Conectividad funcional
Fototest
MoCA
Alzheimer's dementia (AD)
Mild cognitive impairment (MCI)
Electroencephalogram (EEG)
Power spectra
Functional connectivity
dc.subject.proposal.eng.fl_str_mv Alzheimer's dementia (AD)
Mild cognitive impairment (MCI)
Electroencephalogram (EEG)
Power spectra
Functional connectivity
description El abordaje diagnóstico de pacientes con enfermedades que comprometen la cognición es un reto para los sistemas de salud. El electroencefalograma es considerado una técnica útil para evaluar el funcionamiento cerebral, de bajo costo y accesible a la población general. El presente estudio indica que el análisis de la conectividad cerebral y su relación con las pruebas de tamización cognitiva pueden ser un adecuado biomarcador de la función cerebral tanto en las pacientes con enfermedad de Alzheimer como en la población general. Se realizaron análisis tanto en la distribución espectral como en la organización funcional. Se evidenció que el aumento de la potencia Theta se correlacionó negativamente con el desempeño obtenido en las pruebas de tamización cognitiva en toda la población. Por su parte, la conectividad funcional se vio afectada de manera heterogénea. Se demostró una disminución en la sincronización de las oscilaciones Gamma frontotemporales en pacientes con enfermedad de Alzheimer que se correlacionó negativamente con los resultados de las pruebas de tamización. Además, se encontró disminución en la sincronización Theta de similar localización que se correlacionó con la prueba de MOCA. Adicionalmente, se evidenció un aumento en la sincronización Alpha en toda la población que se correlacionó positivamente con los resultados de MOCA.
publishDate 2023
dc.date.issued.none.fl_str_mv 2023
dc.date.accessioned.none.fl_str_mv 2024-02-21T23:56:38Z
dc.date.available.none.fl_str_mv 2024-02-21T23:56:38Z
dc.type.spa.fl_str_mv Trabajo de grado - Doctorado
dc.type.coar.spa.fl_str_mv http://purl.org/coar/resource_type/c_db06
dc.type.content.spa.fl_str_mv Text
dc.type.driver.spa.fl_str_mv info:eu-repo/semantics/doctoralThesis
dc.type.redcol.spa.fl_str_mv http://purl.org/redcol/resource_type/TD
dc.type.version.spa.fl_str_mv info:eu-repo/semantics/draft
dc.type.coarversion.spa.fl_str_mv http://purl.org/coar/version/c_b1a7d7d4d402bcce
format http://purl.org/coar/resource_type/c_db06
status_str draft
dc.identifier.uri.none.fl_str_mv https://hdl.handle.net/11323/10766
dc.identifier.instname.spa.fl_str_mv Corporación Universidad de la Costa
dc.identifier.reponame.spa.fl_str_mv REDICUC-Repositorio.cuc.
dc.identifier.repourl.spa.fl_str_mv https://repositorio.cuc.edu.co/
url https://hdl.handle.net/11323/10766
https://repositorio.cuc.edu.co/
identifier_str_mv Corporación Universidad de la Costa
REDICUC-Repositorio.cuc.
dc.language.iso.spa.fl_str_mv spa
language spa
dc.relation.references.spa.fl_str_mv 2021 Alzheimer’s disease facts and figures. (2021). Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association, 17(3), 327-406. https://doi.org/10.1002/alz.12328
280920-boletines-poblacionales-adulto-mayorI-2020.pdf. (s. f.). Recuperado 15 de marzo de 2022, de https://www.minsalud.gov.co/sites/rid/Lists/BibliotecaDigital/RIDE/DE/PS/280920- boletines-poblacionales-adulto-mayorI-2020.pdf
Adler, G., Brassen, S., & Jajcevic, A. (2003). EEG coherence in Alzheimer’s dementia. Journal of Neural Transmission (Vienna, Austria: 1996), 110(9), 1051-1058. https://doi.org/10.1007/s00702-003-0024-8
Adler, G., Brassen, S., & Jajcevic, A. (2003). EEG coherence in Alzheimer’s dementia. Journal of Neural Transmission (Vienna, Austria: 1996), 110(9), 1051-1058. https://doi.org/10.1007/s00702-003-0024-8
Aggarwal, N. T. (2005). Mild cognitive impairment in different functional domains and incident Alzheimer’s disease. Journal of Neurology, Neurosurgery & Psychiatry, 76(11), 1479-1484. https://doi.org/10.1136/jnnp.2004.053561
Ahmed, R. M., Paterson, R. W., Warren, J. D., Zetterberg, H., O’Brien, J. T., Fox, N. C., Halliday, G. M., & Schott, J. M. (2014). Biomarkers in dementia: Clinical utility and new directions. Journal of Neurology, Neurosurgery & Psychiatry, 85(12), 1426- 1434. https://doi.org/10.1136/jnnp-2014-307662
Albert, M. S., DeKosky, S. T., Dickson, D., Dubois, B., Feldman, H. H., Fox, N. C., Gamst, A., Holtzman, D. M., Jagust, W. J., Petersen, R. C., Snyder, P. J., Carrillo, M. C., Thies, B., & Phelps, C. H. (2011a). The diagnosis of mild cognitive impairment due to Alzheimer’s disease: Recommendations from the National Institute on Aging Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s and Dementia, 7(3), 270-279. Scopus. https://doi.org/10.1016/j.jalz.2011.03.008
Albert, M. S., DeKosky, S. T., Dickson, D., Dubois, B., Feldman, H. H., Fox, N. C., Gamst, A., Holtzman, D. M., Jagust, W. J., Petersen, R. C., Snyder, P. J., Carrillo, M. C., Thies, B., & Phelps, C. H. (2011b). The diagnosis of mild cognitive impairment due to Alzheimer’s disease: Recommendations from the National Institute on Aging Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s & Dementia, 7(3), 270-279. https://doi.org/10.1016/j.jalz.2011.03.008
Al-Nuaimi, A. H. H., Jammeh, E., Sun, L., & Ifeachor, E. (2018). Complexity Measures for Quantifying Changes in Electroencephalogram in Alzheimer’s Disease. Complexity, 2018, e8915079. https://doi.org/10.1155/2018/8915079
Al-Nuaimi, A. H., Jammeh, E., Sun, L., & Ifeachor, E. (2015). Tsallis entropy as a biomarker for detection of Alzheimer’s disease. 2015 37th Annual International Conference of the IEEE Engineering in Medicine and Biology Society (EMBC), 4166-4169.
Al-nuaimi, A. H., Jammeh, E., Sun, L., & Ifeachor, E. (2017). Higuchi fractal dimension of the electroencephalogram as a biomarker for early detection of Alzheimer’s disease. 2017 39th Annual International Conference of the IEEE Engineering in Medicine and Biology Society (EMBC), 2320-2324. https://doi.org/10.1109/EMBC.2017.8037320
Altomare, D., Caprioglio, C., Assal, F., Allali, G., Mendes, A., Ribaldi, F., Ceyzeriat, K., Martins, M., Tomczyk, S., Stampacchia, S., Dodich, A., Boccardi, M., Chicherio, C., Frisoni, G. B., & Garibotto, V. (2021). Diagnostic value of amyloid-PET and tau PET: A head-to-head comparison. European Journal of Nuclear Medicine and Molecular Imaging, 48(7), 2200-2211. Scopus. https://doi.org/10.1007/s00259-021- 05246-x
Alzheimer’s Association. (2014). 2014 Alzheimer’s disease facts and figures. Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association, 10(2), e47-92.
Andersson, E., Janelidze, S., Lampinen, B., Nilsson, M., Leuzy, A., Stomrud, E., Blennow, K., Zetterberg, H., & Hansson, O. (2020). Blood and cerebrospinal fluid neurofilament light differentially detect neurodegeneration in early Alzheimer’s disease. Neurobiology of Aging, 95, 143-153. https://doi.org/10.1016/j.neurobiolaging.2020.07.018
Arango-Lasprilla, J. C., Rivera, D., Garza, M. T., Saracho, C. P., Rodríguez, W., Rodríguez Agudelo, Y., Aguayo, A., Schebela, S., Luna, M., Longoni, M., Martínez, C., Doyle, S., Ocampo-Barba, N., Galarza-del-Angel, J., Aliaga, A., Bringas, M., Esenarro, L., García-Egan, P., & Perrin, P. B. (2015). Hopkins Verbal Learning Test– Revised: Normative data for the Latin American Spanish speaking adult population. NeuroRehabilitation, 37(4), 699-718. https://doi.org/10.3233/NRE-151286
Aron, L., & Yankner, B. A. (2016). Neural synchronization in Alzheimer’s disease. Nature, 540(7632), Art. 7632. https://doi.org/10.1038/540207a
Arráez-Aybar, L.-A., Navia-Álvarez, P., Fuentes-Redondo, T., & Bueno-López, J.-L. (2015). Thomas Willis, a pioneer in translational research in anatomy (on the 350th anniversary of Cerebri anatome). Journal of Anatomy, 226(3), 289-300. https://doi.org/10.1111/joa.12273
Ashburner, J., & Friston, K. J. (2000). Voxel-based morphometry—The methods. NeuroImage, 11(6 Pt 1), 805-821. https://doi.org/10.1006/nimg.2000.0582
Association, A. P. (2014). Guía de consulta de los criterios diagnósticos del DSM-5®: Spanish Edition of the Desk Reference to the Diagnostic Criteria From DSM-5®. American Psychiatric Pub.
Ávila, G. M. R. (2007). Sincronizando con el caos. REBOLDES.
Axmacher, N., & Rasch, B. (2017). Cognitive neuroscience of memory consolidation. Springer.
Babiloni, C., Arakaki, X., Azami, H., Bennys, K., Blinowska, K., Bonanni, L., Bujan, A., Carrillo, M. C., Cichocki, A., de Frutos-Lucas, J., Del Percio, C., Dubois, B., Edelmayer, R., Egan, G., Epelbaum, S., Escudero, J., Evans, A., Farina, F., Fargo, K., … Guntekin, B. (2021). Measures of resting state EEG rhythms for clinical trials in Alzheimer’s disease: Recommendations of an expert panel. Alzheimer’s & Dementia, 17(9), 1528-1553. https://doi.org/10.1002/alz.12311
Babiloni, C., Barry, R. J., Başar, E., Blinowska, K. J., Cichocki, A., Drinkenburg, W. H. I. M., Klimesch, W., Knight, R. T., Lopes da Silva, F., Nunez, P., Oostenveld, R., Jeong, J., Pascual-Marqui, R., Valdes-Sosa, P., & Hallett, M. (2020). International Federation of Clinical Neurophysiology (IFCN) – EEG research workgroup: Recommendations on frequency and topographic analysis of resting state EEG rhythms. Part 1: Applications in clinical research studies. Clinical Neurophysiology, 131(1), 285-307. https://doi.org/10.1016/j.clinph.2019.06.234
Babiloni, C., Binetti, G., Cassarino, A., Dal Forno, G., Del Percio, C., Ferreri, F., Ferri, R., Frisoni, G., Galderisi, S., Hirata, K., Lanuzza, B., Miniussi, C., Mucci, A., Nobili, F., Rodriguez, G., Luca Romani, G., & Rossini, P. M. (2006). Sources of cortical rhythms in adults during physiological aging: A multicentric EEG study. Human Brain Mapping, 27(2), 162-172. https://doi.org/10.1002/hbm.20175
Babiloni, C., Del Percio, C., Caroli, A., Salvatore, E., Nicolai, E., Marzano, N., Lizio, R., Cavedo, E., Landau, S., Chen, K., Jagust, W., Reiman, E., Tedeschi, G., Montella, P., De Stefano, M., Gesualdo, L., Frisoni, G. B., & Soricelli, A. (2016). Cortical sources of resting state EEG rhythms are related to brain hypometabolism in subjects with Alzheimer’s disease: An EEG-PET study. Neurobiology of Aging, 48, 122-134. https://doi.org/10.1016/j.neurobiolaging.2016.08.021
Babiloni, C., Del Percio, C., Lizio, R., Noce, G., Cordone, S., Lopez, S., Soricelli, A., Ferri, R., Pascarelli, M. T., Nobili, F., Arnaldi, D., Famà, F., Aarsland, D., Orzi, F., Buttinelli, C., Giubilei, F., Onofrj, M., Stocchi, F., Stirpe, P., … De Pandis, M. F. (2017). Abnormalities of Cortical Neural Synchronization Mechanisms in Subjects with Mild Cognitive Impairment due to Alzheimer’s and Parkinson’s Diseases: An EEG Study. Journal of Alzheimer’s Disease, 59(1), 339-358. https://doi.org/10.3233/JAD-160883
Babiloni, C., Del Percio, C., Lizio, R., Noce, G., Lopez, S., Soricelli, A., Ferri, R., Pascarelli, M. T., Catania, V., Nobili, F., Arnaldi, D., Famà, F., Orzi, F., Buttinelli, C., Giubilei, F., Bonanni, L., Franciotti, R., Onofrj, M., Stirpe, P., … Stocchi, F. (2018). Functional cortical source connectivity of resting state electroencephalographic alpha rhythms shows similar abnormalities in patients with mild cognitive impairment due to Alzheimer’s and Parkinson’s diseases. Clinical Neurophysiology, 129(4), 766-782. https://doi.org/10.1016/j.clinph.2018.01.009
Babiloni, C., Ferri, R., Binetti, G., Vecchio, F., Frisoni, G. B., Lanuzza, B., Miniussi, C., Nobili, F., Rodriguez, G., Rundo, F., Cassarino, A., Infarinato, F., Cassetta, E., Salinari, S., Eusebi, F., & Rossini, P. M. (2009). Directionality of EEG synchronization in Alzheimer’s disease subjects. Neurobiology of Aging, 30(1), 93- 102. https://doi.org/10.1016/j.neurobiolaging.2007.05.007
Babiloni, C., Frisoni, G. B., Pievani, M., Vecchio, F., Lizio, R., Buttiglione, M., Geroldi, C., Fracassi, C., Eusebi, F., Ferri, R., & Rossini, P. M. (2009). Hippocampal volume and cortical sources of EEG alpha rhythms in mild cognitive impairment and Alzheimer disease. NeuroImage, 44(1), 123-135. https://doi.org/10.1016/j.neuroimage.2008.08.005
Bajo, R., Castellanos, N. P., Cuesta, P., Aurtenetxe, S., Garcia-Prieto, J., Gil-Gregorio, P., del-Pozo, F., & Maestu, F. (2012). Differential Patterns of Connectivity in Progressive Mild Cognitive Impairment. Brain Connectivity, 2(1), 21-24. https://doi.org/10.1089/brain.2011.0069
Balderas, I., Ramírez-Amaya, V., & Bermúdez-Rattoni, F. (2004). [Memory-linked morphological changes]. Revista De Neurologia, 38(10), 944-948.
Barco, A., Alarcon, J. M., & Kandel, E. R. (2002). Expression of constitutively active CREB protein facilitates the late phase of long-term potentiation by enhancing synaptic capture. Cell, 108(5), 689-703.
Barco, A., & Marie, H. (2011). Genetic approaches to investigate the role of CREB in neuronal plasticity and memory. Molecular neurobiology, 44(3), 330-349.
Bartus, R. T., Dean, R. L., Beer, B., & Lippa, A. S. (1982). The cholinergic hypothesis of geriatric memory dysfunction. Science (New York, N.Y.), 217(4558), 408-414. https://doi.org/10.1126/science.7046051
Bateman, R. J., Xiong, C., Benzinger, T. L. S., Fagan, A. M., Goate, A., Fox, N. C., Marcus, D. S., Cairns, N. J., Xie, X., Blazey, T. M., Holtzman, D. M., Santacruz, A., Buckles, V., Oliver, A., Moulder, K., Aisen, P. S., Ghetti, B., Klunk, W. E., McDade, E., … Morris, J. C. (2012). Clinical and Biomarker Changes in Dominantly Inherited Alzheimer’s Disease. New England Journal of Medicine, 367(9), 795-804. https://doi.org/10.1056/NEJMoa1202753
Bauer, R. M., Grande, L., & Valenstein, E. (2003). Amnesic disorders.
Benussi, A., Cantoni, V., Grassi, M., Brechet, L., Michel, C. M., Datta, A., Thomas, C., Gazzina, S., Cotelli, M. S., Bianchi, M., Premi, E., Gadola, Y., Cotelli, M., Pengo, M., Perrone, F., Scolaro, M., Archetti, S., Solje, E., Padovani, A., … Borroni, B. (2022). Increasing Brain Gamma Activity Improves Episodic Memory and Restores Cholinergic Dysfunction in Alzheimer’s Disease. Annals of Neurology, 92(2), 322- 334. Scopus. https://doi.org/10.1002/ana.26411
Besthorn, C., Sattel, H., Geiger-Kabisch, C., Zerfass, R., & Förstl, H. (1995). Parameters of EEG dimensional complexity in Alzheimer’s disease. Electroencephalography and Clinical Neurophysiology, 95(2), 84-89. https://doi.org/10.1016/0013-4694(95)00050- 9
Bethus, I., Tse, D., & Morris, R. G. (2010). Dopamine and memory: Modulation of the persistence of memory for novel hippocampal NMDA receptor-dependent paired associates. Journal of Neuroscience, 30(5), 1610-1618.
Biomarkers Definitions Working Group. (2001). Biomarkers and surrogate endpoints: Preferred definitions and conceptual framework. Clinical Pharmacology and Therapeutics, 69(3), 89-95. https://doi.org/10.1067/mcp.2001.113989
Black, M. M., & Baas, P. W. (1989). The basis of polarity in neurons. Trends in Neurosciences, 12(6), 211-214. https://doi.org/10.1016/0166-2236(89)90124-0
Blennow, K., Dubois, B., Fagan, A. M., Lewczuk, P., de Leon, M. J., & Hampel, H. (2015). Clinical utility of cerebrospinal fluid biomarkers in the diagnosis of early Alzheimer’s disease. Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association, 11(1), 58-69. https://doi.org/10.1016/j.jalz.2014.02.004
Blennow, K., Vanmechelen, E., & Hampel, H. (2001). CSF total tau, Abeta42 and phosphorylated tau protein as biomarkers for Alzheimer’s disease. Molecular Neurobiology, 24(1-3), 87-97. https://doi.org/10.1385/MN:24:1-3:087
Blennow, K., Wallin, A., Agren, H., Spenger, C., Siegfried, J., & Vanmechelen, E. (1995). Tau protein in cerebrospinal fluid: A biochemical marker for axonal degeneration in Alzheimer disease? Molecular and Chemical Neuropathology, 26(3), 231-245. https://doi.org/10.1007/BF02815140
Blennow, K., & Zetterberg, H. (2015). Understanding biomarkers of neurodegeneration: Ultrasensitive detection techniques pave the way for mechanistic understanding. Nature Medicine, 21(3), 217-219. https://doi.org/10.1038/nm.3810
Bliss, T. V., & Collingridge, G. L. (1993). A synaptic model of memory: Long-term potentiation in the hippocampus. Nature, 361(6407), 31-39. https://doi.org/10.1038/361031a0
Bliss, T. V. P., & Cooke, S. F. (2011). Long-term potentiation and long-term depression: A clinical perspective. Clinics, 66(Suppl 1), 3-17. https://doi.org/10.1590/S1807- 59322011001300002
Blom, E. S., Giedraitis, V., Zetterberg, H., Fukumoto, H., Blennow, K., Hyman, B. T., Irizarry, M. C., Wahlund, L.-O., Lannfelt, L., & Ingelsson, M. (2009). Rapid progression from mild cognitive impairment to Alzheimer’s disease in subjects with elevated levels of tau in cerebrospinal fluid and the APOE epsilon4/epsilon4 genotype. Dementia and Geriatric Cognitive Disorders, 27(5), 458-464. https://doi.org/10.1159/000216841
Boada, M., Cejudo, J. C., Tàrraga, L., López, O. L., & Kaufer, D. (2002). Neuropsychiatric Inventory Questionnaire (NPI-Q): Spanish validation of a brief clinical form of the Neuropsychiatric Inventory (NPI). Neurologia, 17(6), 317-323. Scopus.
Bozzali, M., Filippi, M., Magnani, G., Cercignani, M., Franceschi, M., Schiatti, E., Castiglioni, S., Mossini, R., Falautano, M., Scotti, G., Comi, G., & Falini, A. (2006). The contribution of voxel-based morphometry in staging patients with mild cognitive impairment. Neurology, 67(3), 453-460. https://doi.org/10.1212/01.wnl.0000228243.56665.c2
Braak, H., & Braak, E. (1991). Neuropathological stageing of Alzheimer-related changes. Acta Neuropathologica, 82(4), 239-259. https://doi.org/10.1007/BF00308809
Brandt, J. (1991). The Hopkins Verbal Learning Test: Development of a new memory test with six equivalent forms. The clinical neuropsychologist, 5(2), 125-142.
Brandt, J., & Benedict, R. H. (2001). Hopkins verbal learning test—Revised: Professional manual. Psychological Assessment Resources.
Brenner, R. P., Ulrich, R. F., Spiker, D. G., Sclabassi, R. J., Reynolds, C. F., Marin, R. S., & Boller, F. (1986). Computerized EEG spectral analysis in elderly normal, demented and depressed subjects. Electroencephalography and Clinical Neurophysiology, 64(6), 483-492. https://doi.org/10.1016/0013-4694(86)90184-7
Brier, M. R., Thomas, J. B., Snyder, A. Z., Benzinger, T. L., Zhang, D., Raichle, M. E., Holtzman, D. M., Morris, J. C., & Ances, B. M. (2012). Loss of intranetwork and internetwork resting state functional connections with Alzheimer’s disease progression. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, 32(26), 8890-8899. https://doi.org/10.1523/JNEUROSCI.5698- 11.2012
Buckner, R. L. (2012). The serendipitous discovery of the brain’s default network. NeuroImage, 62(2), 1137-1145. https://doi.org/10.1016/j.neuroimage.2011.10.035
Busse, A., Angermeyer, M. C., & Riedel-Heller, S. G. (2006). Progression of mild cognitive impairment to dementia: A challenge to current thinking. The British Journal of Psychiatry: The Journal of Mental Science, 189, 399-404. https://doi.org/10.1192/bjp.bp.105.014779
Buzsáki, G. (2002). Theta Oscillations in the Hippocampus. Neuron, 33(3), 325-340. https://doi.org/10.1016/S0896-6273(02)00586-X
Cabeza, R., & Nyberg, L. (2000). Imaging cognition II: An empirical review of 275 PET and fMRI studies. Journal of Cognitive Neuroscience, 12(1), 1-47. Scopus. https://doi.org/10.1162/08989290051137585
Cacho, J., García-García, R., Arcaya, J., Vicente, J. L., & Lantada, N. (1999). Una propuesta de aplicación y puntuación del test del reloj en la enfermedad de Alzheimer. Rev Neurol, 28(7), 648-655.
Cajal, S. R. (1890). Textura de las circunvolutiones cerebrales de los mamíferos inferiores: Nota preventiva.
Cajal, S. R. Y. (1894). The Croonian Lecture: La Fine Structure des Centres Nerveux. Proceedings of the Royal Society of London, 55(331-335), 444-468. https://doi.org/10.1098/rspl.1894.0063
Cajal, S. R. y. (1899). Textura del sistema nervioso del hombre y de los vertebrados: Estudios sobre el plan estructural y composición histológica de los centros nerviosos adicionados de consideraciones fisiológicas fundadas en los nuevos descubrimentos. N. Moya.
Cajal, S. R. y. (2012). Texture of the Nervous System of Man and the Vertebrates. Springer Science & Business Media.
Cajal, S. R. y, & Cajal, C. S. de I. C. (Spain) I. R. y. (1952). Neuronismo o reticularismo?: Las pruebas objetivas de la unidad anatomica de las celulas nerviosas. Edicion preparada por el Instituto Cajal con motivo del primer centenario de su fundador y maestro.
Camozzato, A. L., Godinho, C., Kochhann, R., Massochini, G., & Chaves, M. L. (2015). Validity of the Brazilian version of the Neuropsychiatric Inventory Questionnaire (NPI-Q). Arquivos de Neuro-Psiquiatria, 73(1), 41-45. Scopus. https://doi.org/10.1590/0004-282X20140177
Can, S. S., Gencay-Can, A., & Gunendi, Z. (2012). Validity and reliability of the clock drawing test as a screening tool for cognitive impairment in patients with fibromyalgia. Comprehensive psychiatry, 53(1), 81-86.
Carnero Pardo, C. (2011). El FOTOTEST en el cribado de demencia en atención primaria. Revista de Neurología, 52(10), 639. https://doi.org/10.33588/rn.5210.2010808
Carnero Pardo, C., Sáez-Zea, C., Montiel Navarro, L., Del Saz, P., Feria Vilar, I., Pérez Navarro, M. J., Ruiz-Giménez, J., Vilchez-Carrillo, R., & Montoro-Rios, M. T. (2007). Diagnostic accuracy of the Phototest for cognitive impairment and dementia. Neurologia, 22(10), 860-869. Scopus.
Carnero-Pardo, C., Sáez-Zea, C., Montiel-Navarro, L., Feria-Vilar, I., & Gurpegui, M. (2011). Estudio normativo y de fiabilidad del FOTOTEST. Neurología, 26(1), 20-25. https://doi.org/10.1016/j.nrl.2010.09.021
Casadio, A., Martin, K. C., Giustetto, M., Zhu, H., Chen, M., Bartsch, D., Bailey, C. H., & Kandel, E. R. (1999). A transient, neuron-wide form of CREB-mediated long-term facilitation can be stabilized at specific synapses by local protein synthesis. Cell, 99(2), 221-237.
Cassani, R., Estarellas, M., San-Martin, R., Fraga, F. J., & Falk, T. H. (2018). Systematic Review on Resting-State EEG for Alzheimer’s Disease Diagnosis and Progression Assessment. Disease Markers, 2018, e5174815. https://doi.org/10.1155/2018/5174815
Casula, E. P., Pellicciari, M. C., Bonnì, S., Borghi, I., Maiella, M., Assogna, M., Minei, M., Motta, C., D’Acunto, A., Porrazzini, F., Pezzopane, V., Mencarelli, L., Roncaioli, A., Rocchi, L., Spampinato, D. A., Caltagirone, C., Santarnecchi, E., Martorana, A., & Koch, G. (2022). Decreased Frontal Gamma Activity in Alzheimer Disease Patients. Annals of Neurology, 92(3), 464-475. Scopus. https://doi.org/10.1002/ana.26444
Cavedo, E., Boccardi, M., Ganzola, R., Canu, E., Beltramello, A., Caltagirone, C., Thompson, P. M., & Frisoni, G. B. (2011). Local amygdala structural differences with 3T MRI in patients with Alzheimer disease. Neurology, 76(8), 727-733. https://doi.org/10.1212/WNL.0b013e31820d62d9
Cecchetti, G., Agosta, F., Basaia, S., Cividini, C., Cursi, M., Santangelo, R., Caso, F., Minicucci, F., Magnani, G., & Filippi, M. (2021). Resting-state electroencephalographic biomarkers of Alzheimer’s disease. NeuroImage: Clinical, 31. Scopus. https://doi.org/10.1016/j.nicl.2021.102711
Cerebri anatome: Cui accessit Nervorum descriptio & usus. (1676). apud Joannem Maximilianum Lucas.
Chandra, A., Dervenoulas, G., Politis, M., & for, the A. D. N. I. (2019). Magnetic resonance imaging in Alzheimer’s disease and mild cognitive impairment. Journal of Neurology, 266(6), 1293-1302. Scopus. https://doi.org/10.1007/s00415-018-9016-3
Chen, A. C., Feng, W., Zhao, H., Yin, Y., & Wang, P. (2008). EEG default mode network in the human brain: Spectral regional field powers. Neuroimage, 41(2), 561-574.
Choi, J., Ku, B., You, Y. G., Jo, M., Kwon, M., Choi, Y., Jung, S., Ryu, S., Park, E., Go, H., Kim, G., Cha, W., & Kim, J. U. (2019). Resting-state prefrontal EEG biomarkers in correlation with MMSE scores in elderly individuals. Scientific Reports, 9(1). Scopus. https://doi.org/10.1038/s41598-019-46789-2
Cieslak, A., Smith, E. E., Lysack, J., & Ismail, Z. (2017). Case series of mild behavioral impairment: Toward an understanding of the early stages of neurodegenerative diseases affecting behavior and cognition. International Psychogeriatrics, 1-8. https://doi.org/10.1017/S1041610217001855
Clute-Reinig, N., Jayadev, S., Rhoads, K., & Le Ny, A.-L. (2021). Alzheimer’s Disease Diagnostics Must Be Globally Accessible. Journal of Alzheimer’s Disease: JAD, 84(4), 1453-1455. https://doi.org/10.3233/JAD-210663
Constitución de Colombia. (1991). ConstitucionColombia.com. https://www.constitucioncolombia.com/titulo-2/capitulo-2/articulo-46
Counter, S. A., Buchanan, L. H., & Ortega, F. (2009). Neurophysiologic and neurocognitive case profiles of andean patients with chronic environmental lead poisoning. Journal of Toxicology and Environmental Health - Part A: Current Issues, 72(19), 1150-1159. Scopus. https://doi.org/10.1080/15287390903091772
Coutanche, M. N., & Thompson-Schill, S. L. (2015). Rapid consolidation of new knowledge in adulthood via fast mapping. Trends in cognitive sciences, 19(9), 486-488
Cowansage, K. K., Shuman, T., Dillingham, B. C., Chang, A., Golshani, P., & Mayford, M. (2014). Direct reactivation of a coherent neocortical memory of context. Neuron, 84(2), 432-441.
Cuesta, P., Ochoa-Urrea, M., Funke, M., Hasan, O., Zhu, P., Marcos, A., López, M. E., Schulz, P. E., Lhatoo, S., Pantazis, D., Mosher, J. C., & Maestu, F. (2022). Gamma band functional connectivity reduction in patients with amnestic mild cognitive impairment and epileptiform activity. Brain Communications, 4(2). Scopus. ¿https://doi.org/10.1093/braincomms/fcac012
Cuesta Prieto, P. (2015). Perfiles biomagnéticos de la actividad en reposo cerebral en las fases preclínica y prodrómica de la Enfermedad de Alzheimer: Influencia del alelo APOE-ε4 y de la acumulación de β-amiloide [Http://purl.org/dc/dcmitype/Text, Universidad Complutense de Madrid]. https://dialnet.unirioja.es/servlet/tesis?codigo=100398
Dauwels, J., Srinivasan, K., Ramasubba Reddy, M., Musha, T., Vialatte, F.-B., Latchoumane, C., Jeong, J., & Cichocki, A. (2011). Slowing and Loss of Complexity in Alzheimer’s EEG: Two Sides of the Same Coin? International Journal of Alzheimer’s Disease, 2011, e539621. https://doi.org/10.4061/2011/539621
Davachi, L., & DuBrow, S. (2015). How the hippocampus preserves order: The role of prediction and context. Trends in cognitive sciences, 19(2), 92-99.
Davis, D. H. J., Creavin, S. T., Yip, J. L. Y., Noel-Storr, A. H., Brayne, C., & Cullum, S. (2021). Montreal Cognitive Assessment for the detection of dementia. Cochrane Database of Systematic Reviews, 2021(7). Scopus. https://doi.org/10.1002/14651858.CD010775.pub3
Davis, H. P., & Squire, L. R. (1984). Protein synthesis and memory: A review. Psychological bulletin, 96(3), 518.
De Carlos, J. A., & Borrell, J. (2007). A historical reflection of the contributions of Cajal and Golgi to the foundations of neuroscience. Brain Research Reviews, 55(1), 8-16. https://doi.org/10.1016/j.brainresrev.2007.03.010
de Frutos-Lucas, J., Cuesta, P., Ramírez-Toraño, F., Nebreda, A., Cuadrado-Soto, E., Peral Suárez, Á., Lopez-Sanz, D., Bruña, R., Marcos-de Pedro, S., Delgado-Losada, M. L., López-Sobaler, A. M., Concepción Rodríguez-Rojo, I., Barabash, A., Serrano Rodriguez, J. M., Laws, S. M., Dolado, A. M., López-Higes, R., Brown, B. M., & Maestú, F. (2020). Age and APOE genotype affect the relationship between objectively measured physical activity and power in the alpha band, a marker of brain disease. Alzheimer’s Research & Therapy, 12, 113. https://doi.org/10.1186/s13195- 020-00681-8
de Jong, L. W., van der Hiele, K., Veer, I. M., Houwing, J. J., Westendorp, R. G. J., Bollen, E. L. E. M., de Bruin, P. W., Middelkoop, H. A. M., van Buchem, M. A., & van der Grond, J. (2008). Strongly reduced volumes of putamen and thalamus in Alzheimer’s disease: An MRI study. Brain, 131(12), 3277-3285. https://doi.org/10.1093/brain/awn278
De La Torre, J. C. (2012). Cardiovascular risk factors promote brain hypoperfusion leading to cognitive decline and dementia. Cardiovascular Psychiatry and Neurology. Scopus. https://doi.org/10.1155/2012/367516
de Sánchez, C. R., Nariño, D., & Cerón, J. F. M. (2010). Epidemiología y carga de la Enfermedad de Alzheimer. Acta Neurol Colomb, 26(3), 1.
DeFelipe, J. (2015a). The dendritic spine story: An intriguing process of discovery. Frontiers in Neuroanatomy, 9, 14. https://doi.org/10.3389/fnana.2015.00014
DeFelipe, J. (2015b). The dendritic spine story: An intriguing process of discovery. Frontiers in Neuroanatomy, 9, 14. https://doi.org/10.3389/fnana.2015.00014
Delbeuck, X., Van der Linden, M., & Collette, F. (2003). Alzheimer’ Disease as a Disconnection Syndrome? Neuropsychology Review, 13(2), 79-92. https://doi.org/10.1023/A:1023832305702
Delgado Derio, C., Guerrero Bonnet, S., Troncoso Ponce, M., Araneda Yañez, A., Slachevsky Chonchol, A., & Behrens Pellegrino, M. I. (2013). Memoria, fluidez y orientación: Prueba de cribado de deterioro cognitivo en 5 minutos. Neurología, 28(7), 400-407. https://doi.org/10.1016/j.nrl.2012.10.001
Demencia. (2020, septiembre 21). https://www.who.int/es/news-room/fact sheets/detail/dementi
Devos, H., Gustafson, K., Liao, K., Ahmadnezhad, P., Estes, B., Martin, L. E., Mahnken, J. D., Brooks, W. M., & Burns, J. M. (2022). EEG/ERP evidence of possible hyperexcitability in older adults with elevated beta-amyloid. Translational Neurodegeneration, 11(1). Scopus. https://doi.org/10.1186/s40035-022-00282-5
Díaz Cabezas, R., Marulanda Mejía, F., Arias, M., & Helena, M. (2013). Prevalence of cognitive impairment and dementia in people older 65 years in a Colombian urban population. Acta Neurológica Colombiana, 29(3), 141-151.
Dierks, T., Jelic, V., Pascual-Marqui, R. D., Wahlund, L.-O., Julin, P., Linden, D. E. J., Maurer, K., Winblad, B., & Nordberg, A. (2000). Spatial pattern of cerebral glucose metabolism (PET) correlates with localization of intracerebral EEG-generators in Alzheimer’s disease. Clinical Neurophysiology, 111(10), 1817-1824. Scopus. https://doi.org/10.1016/S1388-2457(00)00427-2
Dong, Y., Sharma, V. K., Chan, B. P.-L., Venketasubramanian, N., Teoh, H. L., Seet, R. C. S., Tanicala, S., Chan, Y. H., & Chen, C. (2010). The Montreal Cognitive Assessment (MoCA) is superior to the Mini-Mental State Examination (MMSE) for the detection of vascular cognitive impairment after acute stroke. Journal of the Neurological Sciences, 299(1-2), 15-18. https://doi.org/10.1016/j.jns.2010.08.051
Dringenberg, H. C. (2000). Alzheimer’s disease: More than a ‘cholinergic disorder’ —evidence that cholinergic–monoaminergic interactions contribute to EEG slowing and dementia. Behavioural Brain Research, 115(2), 235-249. https://doi.org/10.1016/S0166-4328(00)00261-8
Du, A. T., Schuff, N., Kramer, J. H., Ganzer, S., Zhu, X. P., Jagust, W. J., Miller, B. L., Reed, B. R., Mungas, D., Yaffe, K., Chui, H. C., & Weiner, M. W. (2004). Higher atrophy rate of entorhinal cortex than hippocampus in AD. Neurology, 62(3), 422-427. https://doi.org/10.1212/01.wnl.0000106462.72282.90
Eichenbaum, H. (2011). The Cognitive Neuroscience of Memory: An Introduction. Oxford University Press, USA.
Elwood, R. W. (1991). The Wechsler Memory Scale—Revised: Psychometric characteristics and clinical application. Neuropsychology review, 2(2), 179-201.
Emsell, L., Vanhaute, H., Vansteelandt, K., De Winter, F.-L., Christiaens, D., Van den Stock, J., Vandenberghe, R., Van Laere, K., Sunaert, S., Bouckaert, F., & Vandenbulcke, M. (2022). An optimized MRI and PET based clinical protocol for improving the differential diagnosis of geriatric depression and Alzheimer’s disease. Psychiatry Research - Neuroimaging, 320. Scopus. https://doi.org/10.1016/j.pscychresns.2022.111443
Envejecimiento y salud. (2022, octubre 1). https://www.who.int/es/news-room/fact sheets/detail/ageing-and-healt
Estudio FOTOTRANS: estudio multicéntrico sobre la validez del FOTOTEST en condiciones de práctica clínica | Neurología. (s. f.). Recuperado 10 de marzo de 2022, de https://www.elsevier.es/es-revista-neurologia-295-articulo-estudio-fototrans-estudio multicentrico-sobre-S021348531100260X
Fazekas, F. (1989). Magnetic resonance signal abnormalities in asymptomatic individuals: Their incidence and functional correlates. European Neurology, 29(3), 164-168. https://doi.org/10.1159/000116401
Fonseca, L. C., Tedrus, G. M., Prandi, L. R., Almeida, A. M., & Furlanetto, D. S. (2011). Alzheimer’s disease: Relationship between cognitive aspects and power and coherence EEG measures. Arquivos de Neuro-Psiquiatria, 69(6), 875-881. Scopus. https://doi.org/10.1590/s0004-282x2011000700005
FOTOTEST online. (s. f.). Recuperado 25 de julio de 2022, de https://www.hipocampo.org/FOTOTESTOnline/FOTOTEST-enlinea.asp
Fox, C. A., & Barnard, J. W. (1957). A quantitative study of the Purkinje cell dendritic branchlets and their relationship to afferent fibres. Journal of Anatomy, 91(3), 299- 313.
Frank, D. A., & Greenberg, M. E. (1994). CREB: a mediator of long-term memory from mollusks to mammals. Cell, 79(1), 5-8.
Frey, U., & Morris, R. G. (1998). Synaptic tagging: Implications for late maintenance of hippocampal long-term potentiation. Trends in neurosciences, 21(5), 181-188.
Friston, K. J. (1994). Functional and effective connectivity in neuroimaging: A synthesis. Human Brain Mapping, 2(1-2), 56-78. https://doi.org/10.1002/hbm.460020107
Fuchs, M., Kastner, J., Wagner, M., Hawes, S., & Ebersole, J. S. (2002). A standardized boundary element method volume conductor model. Clinical Neurophysiology: Official Journal of the International Federation of Clinical Neurophysiology, 113(5), 702-712. https://doi.org/10.1016/s1388-2457(02)00030-5
Gallistel, C. R., & Matzel, L. D. (2013). The neuroscience of learning: Beyond the Hebbian synapse. Annual Review of Psychology, 64, 169-200. https://doi.org/10.1146/annurev psych-113011-143807
Ganguli, M., Dodge, H. H., Shen, C., & DeKosky, S. T. (2004). Mild cognitive impairment, amnestic type: An epidemiologic study. Neurology, 63(1), 115-121.
García-Morales, P., Gich-Fullà, J., Guardia-Olmos, J., & Casanova, J. P. (1998). Series de dígitos, series automáticas y orientación: Normas ampliadas del Test Barcelona. Neurología (Barc., Ed. impr.), 13(S-6), 277-286.
García-Sánchez, J. E., García, E., & Lucila Merino, M. (2010). Cien años de la bala mágica del Dr. Ehrlich (1909–2009). Enfermedades Infecciosas y Microbiología Clínica, 28(8), 521-533. https://doi.org/10.1016/j.eimc.2009.07.009
Gasser, T., & Molinari, L. (1996). The analysis of the EEG. Statistical Methods in Medical Research, 5(1), 67-99. https://doi.org/10.1177/096228029600500105
Ge, Q., Lin, Z.-C., Gao, Y.-X., & Zhang, J.-X. (2020). A Robust Discriminant Framework Based on Functional Biomarkers of EEG and Its Potential for Diagnosis of Alzheimer’s Disease. Healthcare, 8(4), Art. 4. https://doi.org/10.3390/healthcare8040476
Gianotti, L. R. R., Künig, G., Lehmann, D., Faber, P. L., Pascual-Marqui, R. D., Kochi, K., & Schreiter-Gasser, U. (2007). Correlation between disease severity and brain electric LORETA tomography in Alzheimer’s disease. Clinical Neurophysiology: Official Journal of the International Federation of Clinical Neurophysiology, 118(1), 186- 196. https://doi.org/10.1016/j.clinph.2006.09.007
Gili, T., Cercignani, M., Serra, L., Perri, R., Giove, F., Maraviglia, B., Caltagirone, C., & Bozzali, M. (2011). Regional brain atrophy and functional disconnection across Alzheimer’s disease evolution. Journal of Neurology, Neurosurgery, and Psychiatry, 82(1), 58-66. https://doi.org/10.1136/jnnp.2009.199935
Golby, A., Silverberg, G., Race, E., Gabrieli, S., O’Shea, J., Knierim, K., Stebbins, G., & Gabrieli, J. (2005). Memory encoding in Alzheimer’s disease: An fMRI study of explicit and implicit memory. Brain: A Journal of Neurology, 128(Pt 4), 773-787. https://doi.org/10.1093/brain/awh40
Gómez, F. R., Guerrero, C. E. D., Bernal, G. B., Pongutá, J. P. C., & Olarte, J. L. R. (s. f.). Politica colombiana de envejecimiento humano y vejez 2015—2024. 54.
Gonzalez-Escamilla, G., Atienza, M., & Cantero, J. L. (2015). Impaired cortical oscillatory coupling in mild cognitive impairment: Anatomical substrate and ApoE4 effects. Brain Structure & Function, 220(3), 1721-1737. https://doi.org/10.1007/s00429-014-0757-1
Gordon, B. A., Najmi, S., Hsu, P., Roe, C. M., Morris, J. C., & Benzinger, T. L. (2015). The effects of white matter hyperintensities and amyloid deposition on Alzheimer dementia. NeuroImage: Clinical, 8, 246-252.
Gouveia, P. A. R., Brucki, S. M. D., Malheiros, S. M. F., & Bueno, O. F. A. (2007). Disorders in planning and strategy application in frontal lobe lesion patients. Brain and cognition, 63(3), 240-246.
Gray, E. G. (1959). Electron Microscopy of Synaptic Contacts on Dendrite Spines of the Cerebral Cortex. Nature, 183(4675), 1592-1593. https://doi.org/10.1038/1831592a0
Greicius, M. D., Krasnow, B., Reiss, A. L., & Menon, V. (2003). Functional connectivity in the resting brain: A network analysis of the default mode hypothesis. Proceedings of the National Academy of Sciences, 100(1), 253-258. https://doi.org/10.1073/pnas.0135058100
Greicius, M. D., Srivastava, G., Reiss, A. L., & Menon, V. (2004). Default-mode network activity distinguishes Alzheimer’s disease from healthy aging: Evidence from functional MRI. Proceedings of the National Academy of Sciences of the United States of America, 101(13), 4637-4642. https://doi.org/10.1073/pnas.0308627101
Gu, F., Meng, X., Shen, E., & Cai, Z. (2003). Can we measure consciousness with eeg complexities? International Journal of Bifurcation and Chaos, 13(03), 733-742. https://doi.org/10.1142/S0218127403006893
Hall, B., Mak, E., Cervenka, S., Aigbirhio, F. I., Rowe, J. B., & O’Brien, J. T. (2017). In vivo tau PET imaging in dementia: Pathophysiology, radiotracer quantification, and a systematic review of clinical findings. Ageing Research Reviews, 36, 50-63. Scopus. https://doi.org/10.1016/j.arr.2017.03.002
Handayani, N., Haryanto, F., Khotimah, S. N., Arif, I., & Taruno, W. P. (2018). Coherence and phase synchrony analyses of EEG signals in Mild Cognitive Impairment (MCI): A study of functional brain connectivity. Polish Journal of Medical Physics and Engineering, 24(1), 1-9. Scopus. https://doi.org/10.2478/pjmpe-2018-0001
Hanslmayr, S., & Staudigl, T. (2014). How brain oscillations form memories—A processing based perspective on oscillatory subsequent memory effects. NeuroImage, 85, 648-655. https://doi.org/10.1016/j.neuroimage.2013.05.121
Hebb, D. O. (1949). The organization of behavior: A neuropsychological approach. John Wiley & Sons.
Hemmy, L. S., Linskens, E. J., Silverman, P. C., Miller, M. A., Talley, K. M. C., Taylor, B. C., Ouellette, J. M., Greer, N. L., Wilt, T. J., Butler, M., & Fink, H. A. (2020). Brief cognitive tests for distinguishing clinical Alzheimer-type dementia from mild cognitive impairment or normal cognition in older adults with suspected cognitive impairment: A systematic review. Annals of Internal Medicine, 172(10), 678-687. Scopus. https://doi.org/10.7326/M19-3889
Henry, J. D., & Crawford, J. R. (2004). A meta-analytic review of verbal fluency performance in patients with traumatic brain injury. Neuropsychology, 18(4), 621.
Horvath, A. (2018). EEG and ERP biomarkers of Alzheimer rsquo s disease a critical review. Frontiers in Bioscience, 23(1), 183-220. https://doi.org/10.2741/4587
Hsu, Y.-H., Huang, S.-M., Lin, S.-Y., Yang, J.-J., Tu, M.-C., & Kuo, L.-W. (2022). Prospective Memory and Default Mode Network Functional Connectivity in Normal and Pathological Aging. Journal of Alzheimer’s Disease, 86(2), 753-762. Scopus. https://doi.org/10.3233/JAD-215293
Huang, Y. Y., & Kandel, E. R. (1994). Recruitment of long-lasting and protein kinase A dependent long-term potentiation in the CA1 region of hippocampus requires repeated tetanization. Learning & Memory, 1(1), 74-82. https://doi.org/10.1101/lm.1.1.74
Huey, E. D., Manly, J. J., Tang, M.-X., Schupf, N., Brickman, A. M., Manoochehri, M., Mez, J., DeCarli, C., Devanand, D. P., & Mayeux, R. (2013). Course and etiology of dysexecutive MCI in a community sample. Alzheimer’s & dementia : the journal of the Alzheimer’s Association, 9(6), 632-639. https://doi.org/10.1016/j.jalz.2012.10.014
Hunt, K., Adamson, J., Hewitt, C., & Nazareth, I. (2011). Do women consult more than men? A review of gender and consultation for back pain and headache. Journal of Health Services Research and Policy, 16(2), 108-117. Scopus. https://doi.org/10.1258/jhsrp.2010.009131
Huygens, C. (1673). Horologium Oscillatorium.
Iaccarino, H. F., Singer, A. C., Martorell, A. J., Rudenko, A., Gao, F., Gillingham, T. Z., Mathys, H., Seo, J., Kritskiy, O., Abdurrob, F., Adaikkan, C., Canter, R. G., Rueda, R., Brown, E. N., Boyden, E. S., & Tsai, L.-H. (2016). Gamma frequency entrainment attenuates amyloid load and modifies microglia. Nature, 540(7632), Art. 7632. https://doi.org/10.1038/nature20587
Ibanez, A., Parra, M. A., Butler, C., & Latin America and the Caribbean Consortium on Dementia (LAC-CD). (2021). The Latin America and the Caribbean Consortium on Dementia (LAC-CD): From Networking to Research to Implementation Science. Journal of Alzheimer’s Disease: JAD, 82(s1), S379-S394. https://doi.org/10.3233/JAD-201384
Isaac, J. T., Nicoll, R. A., & Malenka, R. C. (1995). Evidence for silent synapses: Implications for the expression of LTP. Neuron, 15(2), 427-434.
Ivan Pavlov—Biographical. (s. f.). Recuperado 4 de noviembre de 2017, de https://www.nobelprize.org/nobel_prizes/medicine/laureates/1904/pavlov-bio.html
Jack, C. R., Bennett, D. A., Blennow, K., Carrillo, M. C., Dunn, B., Haeberlein, S. B., Holtzman, D. M., Jagust, W., Jessen, F., Karlawish, J., Liu, E., Molinuevo, J. L., Montine, T., Phelps, C., Rankin, K. P., Rowe, C. C., Scheltens, P., Siemers, E., Snyder, H. M., … Contributors. (2018). NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association, 14(4), 535-562. https://doi.org/10.1016/j.jalz.2018.02.018
Janelidze, S., Stomrud, E., Smith, R., Palmqvist, S., Mattsson, N., Airey, D. C., Proctor, N. K., Chai, X., Shcherbinin, S., Sims, J. R., Triana-Baltzer, G., Theunis, C., Slemmon, R., Mercken, M., Kolb, H., Dage, J. L., & Hansson, O. (2020). Cerebrospinal fluid p-tau217 performs better than p-tau181 as a biomarker of Alzheimer’s disease. Nature Communications, 11(1), 1683. https://doi.org/10.1038/s41467-020-15436-0
Jarrett, J. T., Berger, E. P., & Lansbury, P. T. (1993). The carboxy terminus of the beta amyloid protein is critical for the seeding of amyloid formation: Implications for the pathogenesis of Alzheimer’s disease. Biochemistry, 32(18), 4693-4697. https://doi.org/10.1021/bi00069a001
Jelic, V., Johansson, S.-E., Almkvist, O., Shigeta, M., Julin, P., Nordberg, A., Winblad, B., & Wahlund, L.-O. (2000). Quantitative electroencephalography in mild cognitive impairment: Longitudinal changes and possible prediction of Alzheimer’s disease. Neurobiology of Aging, 21(4), 533-540. https://doi.org/10.1016/S0197-4580(00)00153-6
Jeong, J., Gore, J. C., & Peterson, B. S. (2001). Mutual information analysis of the EEG in patients with Alzheimer’s disease. Clinical Neurophysiology, 112(5), 827-835. https://doi.org/10.1016/S1388-2457(01)00513-2
Johnson, N., Barion, A., Rademaker, A., Rehkemper, G., & Weintraub, S. (2004). The Activities of Daily Living Questionnaire: A validation study in patients with dementia. Alzheimer disease & associated disorders, 18(4), 223-230.
Jongsiriyanyong, S., & Limpawattana, P. (2018). Mild Cognitive Impairment in Clinical Practice: A Review Article. American Journal of Alzheimer’s Disease and Other Dementias, 33(8), 500-507. Scopus. https://doi.org/10.1177/1533317518791401
Josselyn, S. A., Köhler, S., & Frankland, P. W. (2015). Finding the engram. Nature Reviews Neuroscience, 16(9), 521-534. https://doi.org/10.1038/nrn4000
Kandel, E. R. (2007). En busca de la memoria: El nacimiento de una nueva ciencia de la mente. Katz Editores.
Kandel, E. R., Schwartz, J. H., & Jessell, T. M. (2000). Principios de neurociencia. McGraw Hill Interamericana
Kandel, E. R., & Spencer, W. A. (1968). Cellular neurophysiological approaches in the study of learning. Physiological Reviews, 48(1), 65-134.
Kapasi, A., DeCarli, C., & Schneider, J. A. (2017). Impact of multiple pathologies on the threshold for clinically overt dementia. Acta Neuropathologica, 134(2), 171-186. https://doi.org/10.1007/s00401-017-1717-7
Karikari, T. K., Benedet, A. L., Ashton, N. J., Lantero Rodriguez, J., Snellman, A., Suárez Calvet, M., Saha-Chaudhuri, P., Lussier, F., Kvartsberg, H., Rial, A. M., Pascoal, T. A., Andreasson, U., Schöll, M., Weiner, M. W., Rosa-Neto, P., Trojanowski, J. Q., Shaw, L. M., Blennow, K., Zetterberg, H., & Alzheimer’s Disease Neuroimaging Initiative. (2021). Diagnostic performance and prediction of clinical progression of plasma phospho-tau181 in the Alzheimer’s Disease Neuroimaging Initiative. Molecular Psychiatry, 26(2), 429-442. https://doi.org/10.1038/s41380-020-00923-z
Karikari, T. K., Pascoal, T. A., Ashton, N. J., Janelidze, S., Benedet, A. L., Rodriguez, J. L., Chamoun, M., Savard, M., Kang, M. S., Therriault, J., Schöll, M., Massarweh, G., Soucy, J.-P., Höglund, K., Brinkmalm, G., Mattsson, N., Palmqvist, S., Gauthier, S., Stomrud, E., … Blennow, K. (2020). Blood phosphorylated tau 181 as a biomarker for Alzheimer’s disease: A diagnostic performance and prediction modelling study using data from four prospective cohorts. The Lancet. Neurology, 19(5), 422-433. https://doi.org/10.1016/S1474-4422(20)30071-5
Kauer, J. A., Malenka, R. C., & Nicoll, R. A. (1988). NMDA application potentiates synaptic transmission in the hippocampus. Nature, 334(6179), 250-252. https://doi.org/10.1038/334250a0
Kichigina, V. F. (2019). Disturbances of theta and gamma oscillations in the brains with pathologies pecular to Alzheimer’s disease and temporal lobe epilepsy. Zhurnal Vysshei Nervnoi Deyatelnosti Imeni I.P. Pavlova, 69(4), 395-412. Scopus. https://doi.org/10.1134/S0044467719040075
Klyucherev, T. O., Olszewski, P., Shalimova, A. A., Chubarev, V. N., Tarasov, V. V., Attwood, M. M., Syvänen, S., & Schiöth, H. B. (2022). Advances in the development of new biomarkers for Alzheimer’s disease. Translational Neurodegeneration, 11(1). Scopus. https://doi.org/10.1186/s40035-022-00296-z
Koenig, T., Prichep, L., Dierks, T., Hubl, D., Wahlund, L. O., John, E. R., & Jelic, V. (2005). Decreased EEG synchronization in Alzheimer’s disease and mild cognitive impairment. Neurobiology of Aging, 26(2), 165-171. https://doi.org/10.1016/j.neurobiolaging.2004.03.008
Kölliker, A., & Kölliker, A. von. (1863). Handbuch der Gewebelehre des Menschen: Für Aerzte und Studirende. Wilhelm Engelmann.
Kowalski, J. W., Gawel, M., Pfeffer, A., & Barcikowska, M. (2001). The Diagnostic Value of EEG in Alzheimer Disease: Correlation With the Severity of Mental Impairment. Journal of Clinical Neurophysiology, 18(6), 570-575.
Kulkarni, N., & Bairagi, V. (2018). Chapter Two—Electroencephalogram and Its Use in Clinical Neuroscience. En N. Kulkarni & V. Bairagi (Eds.), EEG-Based Diagnosis of Alzheimer Disease (pp. 25-35). Academic Press. https://doi.org/10.1016/B978-0-12-815392-5.00002-2
Kwak, Y. T. (2006). Quantitative EEG findings in different stages of Alzheimer’s disease. Journal of Clinical Neurophysiology: Official Publication of the American Electroencephalographic Society, 23(5), 456-461. https://doi.org/10.1097/01.wnp.0000223453.47663.63
Lamar, M., Boots, E. A., Arfanakis, K., Barnes, L. L., & Schneider, J. A. (2020). Common Brain Structural Alterations Associated with Cardiovascular Disease Risk Factors and Alzheimer’s Dementia: Future Directions and Implications. Neuropsychology Review, 30(4), 546-557. Scopus. https://doi.org/10.1007/s11065-020-09460-6
Larson, J., & Munkácsy, E. (2015). Theta-Burst LTP. Brain research, 1621, 38-50. https://doi.org/10.1016/j.brainres.2014.10.034
Lashley, K. S. (1950). In search of the engram.
Laufs, H. (2008). Endogenous brain oscillations and related networks detected by surface EEG‐combined fMRI. Human brain mapping, 29(7), 762-769.
Lee, S.-H., Park, Y.-M., Kim, D.-W., & Im, C.-H. (2010). Global synchronization index as a biological correlate of cognitive decline in Alzheimer’s disease. Neuroscience Research, 66(4), 333-339. Scopus. https://doi.org/10.1016/j.neures.2009.12.004
Lempel, A., & Ziv, J. (1976). On the Complexity of Finite Sequences. IEEE Transactions on Information Theory, 22(1), 75-81. https://doi.org/10.1109/TIT.1976.1055501
Lenzi, D., Serra, L., Perri, R., Pantano, P., Lenzi, G. L., Paulesu, E., Caltagirone, C., Bozzali, M., & Macaluso, E. (2011). Single domain amnestic MCI: A multiple cognitive domains fMRI investigation. Neurobiology of Aging, 32(9), 1542-1557. https://doi.org/10.1016/j.neurobiolaging.2009.09.006
Leuzy, A., Cullen, N. C., Mattsson-Carlgren, N., & Hansson, O. (2021). Current advances in plasma and cerebrospinal fluid biomarkers in Alzheimer’s disease. Current Opinion in Neurology, 34(2), 266-274. https://doi.org/10.1097/WCO.0000000000000904
Ley 1850 de 2017—Medidas de protección al adulto mayor en Colombia. (2018, mayo 10). Así Vamos en Salud - indicadores en salud normatividad derechos. https://www.asivamosensalud.org/politicas-publicas/normatividad-leyes/promocion social/ley-1850-de-2017-medidas-de-proteccion-al
Li, C., Zheng, J., Wang, J., Gui, L., & Li, C. (2009). An fMRI stroop task study of prefrontal cortical function in normal aging, mild cognitive impairment, and Alzheimer’s disease. Current Alzheimer Research, 6(6), 525-530. https://doi.org/10.2174/156720509790147142
Lizio, R., Del Percio, C., Marzano, N., Soricelli, A., Yener, G. G., Başar, E., Mundi, C., De Rosa, S., Triggiani, A. I., Ferri, R., Arnaldi, D., Nobili, F. M., Cordone, S., Lopez, S., Carducci, F., Santi, G., Gesualdo, L., Rossini, P. M., Cavedo, E., … Babiloni, C. (2016). Neurophysiological assessment of Alzheimer’s disease individuals by a single electroencephalographic marker. Journal of Alzheimer’s Disease: JAD, 49(1), 159-177. https://doi.org/10.3233/JAD-143042
Lizio, R., Vecchio, F., Frisoni, G. B., Ferri, R., Rodriguez, G., & Babiloni, C. (2011). Electroencephalographic rhythms in Alzheimer’s disease. International Journal of Alzheimer’s Disease, 2011, 927573. https://doi.org/10.4061/2011/927573
Lleó, A., Alcolea, D., Martínez-Lage, P., Scheltens, P., Parnetti, L., Poirier, J., Simonsen, A. H., Verbeek, M. M., Rosa-Neto, P., Slot, R. E. R., Tainta, M., Izaguirre, A., Reijs, B. L. R., Farotti, L., Tsolaki, M., Vandenbergue, R., Freund-Levi, Y., Verhey, F. R. J., Clarimón, J., … Blennow, K. (2019). Longitudinal cerebrospinal fluid biomarker trajectories along the Alzheimer’s disease continuum in the BIOMARKAPD study. Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association, 15(6), 742-753. https://doi.org/10.1016/j.jalz.2019.01.015
Locatelli, T., Cursi, M., Liberati, D., Franceschi, M., & Comi, G. (1998). EEG coherence in Alzheimer’s disease. Electroencephalography and Clinical Neurophysiology, 106(3), 229-237. https://doi.org/10.1016/S0013-4694(97)00129-6
Lomo, T. (1966). Frequency potentiation of excitatory synaptic activity in dentate area of hippocampal formation. Acta Physiologica Scandinavica, 128-.
Lomo, T. (1971). Patterns of activation in a monosynaptic cortical pathway: The perforant path input to the dentate area of the hippocampal formation. Experimental Brain Research, 12(1), 18-45.
Lømo, T. (2003). The discovery of long-term potentiation. Philosophical Transactions of the Royal Society B: Biological Sciences, 358(1432), 617-620. https://doi.org/10.1098/rstb.2002.1226
López, N., Allegri, R., & Soto-Añari, M. (2014). Capacidad diagnóstica y validación preliminar del Test del Reloj, versión de Cacho a la Orden, para enfermedad de Alzheimer de grado leve en población chilena. Revista Ecuatoriana de Neurología, 23(1-3), 18-23.
Loureiro, C. C. de S., García, C., Adana, L., Yacelga, T., Rodríguez Lorenzana, A., & Maruta, C. (2018). Uso del test de evaluación cognitiva de Montreal (MoCA) en América Latina: Revisión sistemática. Revista de Neurología, 66(12), 397. https://doi.org/10.33588/rn.6612.2017508
Lu, W., Man, H., Ju, W., Trimble, W. S., MacDonald, J. F., & Wang, Y. T. (2001). Activation of synaptic NMDA receptors induces membrane insertion of new AMPA receptors and LTP in cultured hippocampal neurons. Neuron, 29(1), 243-254.
Lüscher, C., & Frerking, M. (2001). Restless AMPA receptors: Implications for synaptic transmission and plasticity. Trends in Neurosciences, 24(11), 665-670
Lynch, M. A. (2004). Long-term potentiation and memory. Physiological Reviews, 84(1), 87-136. https://doi.org/10.1152/physrev.00014.2003
Maestú, F., Cuesta, P., Hasan, O., Fernandéz, A., Funke, M., & Schulz, P. E. (2019). The Importance of the Validation of M/Eeg with Current Biomarkers in Alzheimer’s Disease. Frontiers in Human Neuroscience, 13. https://www.frontiersin.org/article/10.3389/fnhum.2019.00017
Malenka, R. C., Kauer, J. A., Zucker, R. S., & Nicoll, R. A. (1988). Postsynaptic calcium is sufficient for potentiation of hippocampal synaptic transmission. Science, 242(4875), 81-84. https://doi.org/10.1126/science.2845577
Malinow, R., & Malenka, R. C. (2002). AMPA receptor trafficking and synaptic plasticity. Annual Review of Neuroscience, 25, 103-126. https://doi.org/10.1146/annurev.neuro.25.112701.142758
Mallo, S. C., Patten, S. B., Ismail, Z., Pereiro, A. X., Facal, D., Otero, C., & Juncos-Rabadán, O. (2020). Does the neuropsychiatric inventory predict progression from mild cognitive impairment to dementia? A systematic review and meta-analysis. Ageing Research Reviews, 58. Scopus. https://doi.org/10.1016/j.arr.2019.101004
Manning, J. R., Polyn, S. M., Baltuch, G. H., Litt, B., & Kahana, M. J. (2011). Oscillatory patterns in temporal lobe reveal context reinstatement during memory search. Proceedings of the National Academy of Sciences, 108(31), 12893-12897.
Matsunari, I. (2012). MRI and PET in Alzheimer’s disease. Neurological Surgery, 40(12), 1051-1063. Scopus.
Mattia, D., Babiloni, F., Romigi, A., Cincotti, F., Bianchi, L., Sperli, F., Placidi, F., Bozzao, A., Giacomini, P., Floris, R., & Marciani, M. G. (2003). Quantitative EEG and dynamic susceptibility contrast MRI in Alzheimer’s disease: A correlative study. Clinical Neurophysiology, 114(7), 1210-1216. Scopus. https://doi.org/10.1016/S1388-2457(03)00085-3
Mazziotta, J. C., Toga, A. W., Evans, A., Fox, P., & Lancaster, J. (1995). A probabilistic atlas of the human brain: Theory and rationale for its development. The International Consortium for Brain Mapping (ICBM). NeuroImage, 2(2), 89-101. https://doi.org/10.1006/nimg.1995.1012
McGeown, W. J., Shanks, M. F., Forbes-McKay, K. E., & Venneri, A. (2009). Patterns of brain activity during a semantic task differentiate normal aging from early Alzheimer’s disease. Psychiatry Research, 173(3), 218-227. https://doi.org/10.1016/j.pscychresns.2008.10.005
McKhann, G. M., Knopman, D. S., Chertkow, H., Hyman, B. T., Jack, C. R., Kawas, C. H., Klunk, W. E., Koroshetz, W. J., Manly, J. J., Mayeux, R., Mohs, R. C., Morris, J. C., Rossor, M. N., Scheltens, P., Carrillo, M. C., Thies, B., Weintraub, S., & Phelps, C. H. (2011). The diagnosis of dementia due to Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s & Dementia, 7(3), 263-269. https://doi.org/10.1016/j.jalz.2011.03.005
Miledi, R., & Slater, C. R. (1966). The action of calcium on neuronal synapses in the squid. The Journal of Physiology, 184(2), 473-498. https://doi.org/10.1113/jphysiol.1966.sp007927
MINISTERIO DE SALUD Y PROTECCIÓN SOCIAL. (2013). Envejecimiento demografico-Colombia-1951-2020. OFICINA DE PROMOCIÓN SOCIAL. https://www.minsalud.gov.co/sites/rid/Lists/BibliotecaDigital/RIDE/DE/PS/Envejecimiento-demografico-Colombia-1951-2020.pd
Möller, C., Van Der Flier, W. M., Versteeg, A., Benedictus, M. R., Wattjes, M. P., Koedam, E. L. G. M., Scheltens, P., Barkhof, F., & Vrenken, H. (2014). Quantitative regional validation of the visual rating scale for posterior cortical atrophy. European Radiology, 24(2), 397-404. Scopus. https://doi.org/10.1007/s00330-013-3025-5
Monllor, P., Cervera-Ferri, A., Lloret, M.-A., Esteve, D., Lopez, B., Leon, J.-L., & Lloret, A. (2021). Electroencephalography as a non-invasive biomarker of alzheimer’s disease: A forgotten candidate to substitute CSF molecules? International Journal of Molecular Sciences, 22(19). Scopus. https://doi.org/10.3390/ijms221910889
Montez, T., Linkenkaer-Hansen, K., van Dijk, B. W., & Stam, C. J. (2006). Synchronization likelihood with explicit time-frequency priors. NeuroImage, 33(4), 1117-1125. https://doi.org/10.1016/j.neuroimage.2006.06.066
Moretti, D. V. (2015). Theta and alpha eeg frequency interplay in subjects with mild cognitive impairment: Evidence from EEG, MRI and spect brain modifications. Frontiers in Aging Neuroscience, 7(FEB). Scopus. https://doi.org/10.3389/fnagi.2015.00031
Morris, J. C. (2006). Mild Cognitive Impairment Is Early-Stage Alzheimer Disease: Time to Revise Diagnostic Criteria. Archives of Neurology, 63(1), 15-16. https://doi.org/10.1001/archneur.63.1.15
Morris, R. G., Anderson, E., Lynch, G. S., & Baudry, M. (1986). Selective impairment of learning and blockade of long-term potentiation by an N-methyl-D-aspartate receptor antagonist, AP5. Nature, 319(6056), 774-776. https://doi.org/10.1038/319774a0
Morris, R. G. M. (2006). Elements of a neurobiological theory of hippocampal function: The role of synaptic plasticity, synaptic tagging and schemas. European Journal of Neuroscience, 23(11), 2829-2846. https://doi.org/10.1111/j.1460-9568.2006.04888.x
Morris, R. G. M. (2013). NMDA receptors and memory encoding. Neuropharmacology, 74, 32-40. https://doi.org/10.1016/j.neuropharm.2013.04.014
Mullany, P., & Lynch, M. A. (1997). Changes in protein synthesis and synthesis of the synaptic vesicle protein, synaptophysin, in entorhinal cortex following induction of long-term potentiation in dentate gyrus: An age-related study in the rat. Neuropharmacology, 36(7), 973-980.
Muñoz-Neira, C., López, O. L., Riveros, R., Núñez-Huasaf, J., Flores, P., & Slachevsky, A. (2012). The Technology – Activities of Daily Living Questionnaire: A Version with a Technology-Related Subscale. Dementia and Geriatric Cognitive Disorders, 33(6), 361-371. https://doi.org/10.1159/000338606
Murphy, D. D., & Segal, M. (1997). Morphological plasticity of dendritic spines in central neurons is mediated by activation of cAMP response element binding protein. Proceedings of the National Academy of Sciences, 94(4), 1482-1487.
Musa, G., Henríquez, F., Muñoz-Neira, C., Delgado, C., Lillo, P., & Slachevsky, A. (2017). Utility of the Neuropsychiatric Inventory Questionnaire (NPI-Q) in the assessment of a sample of patients with Alzheimer’s disease in Chile. Dementia & Neuropsychologia, 11(2), 129-136. https://doi.org/10.1590/1980-57642016dn11- 020005
Musaeus, C. S., Engedal, K., Høgh, P., Jelic, V., Mørup, M., Naik, M., Oeksengaard, A.-R., Snaedal, J., Wahlund, L.-O., Waldemar, G., & Andersen, B. B. (2018). EEG Theta Power Is an Early Marker of Cognitive Decline in Dementia due to Alzheimer’s Disease. Journal of Alzheimer’s Disease, 64(4), 1359-1371. https://doi.org/10.3233/JAD-180300
Musaeus, C. S., Engedal, K., Høgh, P., Jelic, V., Mørup, M., Naik, M., Oeksengaard, A.-R., Snaedal, J., Wahlund, L.-O., Waldemar, G., & Andersen, B. B. (2019). Oscillatory connectivity as a diagnostic marker of dementia due to Alzheimer’s disease. Clinical Neurophysiology, 130(10), 1889-1899.https://doi.org/10.1016/j.clinph.2019.07.016
Nakamura, A., Cuesta, P., Fernández, A., Arahata, Y., Iwata, K., Kuratsubo, I., Bundo, M., Hattori, H., Sakurai, T., Fukuda, K., Washimi, Y., Endo, H., Takeda, A., Diers, K., Bajo, R., Maestú, F., Ito, K., & Kato, T. (2018). Electromagnetic signatures of the preclinical and prodromal stages of Alzheimer’s disease. Brain, 141(5), 1470-1485. https://doi.org/10.1093/brain/awy044
Nakamura, A., Kaneko, N., Villemagne, V. L., Kato, T., Doecke, J., Doré, V., Fowler, C., Li, Q.-X., Martins, R., Rowe, C., Tomita, T., Matsuzaki, K., Ishii, K., Ishii, K., Arahata, Y., Iwamoto, S., Ito, K., Tanaka, K., Masters, C. L., & Yanagisawa, K. (2018). High performance plasma amyloid-β biomarkers for Alzheimer’s disease. Nature, 554(7691), 249-254. https://doi.org/10.1038/nature25456
Nasreddine, Z. S., Phillips, N. A., Bédirian, V., Charbonneau, S., Whitehead, V., Collin, I., Cummings, J. L., & Chertkow, H. (2005). The Montreal Cognitive Assessment, MoCA: A Brief Screening Tool For Mild Cognitive Impairment. Journal of the American Geriatrics Society, 53(4), 695-699. https://doi.org/10.1111/j.1532- 5415.2005.53221.x
Natham, R., Amirthalingam, P., & Arunachalam, G. (2018). Comparison of Montreal Cognitive Assessment (MOCA) with Mini Mental State Examination (MMSE) on association between homocysteine and cognitive status in epilepsy patients with phenytoin monotherapy. Asia Pacific Journal of Counselling and Psychotherapy, 9(2), 160-170. https://doi.org/10.1080/21507686.2018.1483411
Nesteruk, M., Nesteruk, T., Styczyńska, M., Barczak, A., Mandecka, M., Walecki, J., & Barcikowska-Kotowicz, M. (2015). Predicting the conversion of mild cognitive impairment to Alzheimer’s disease based on the volumetric measurements of the selected brain structures in magnetic resonance imaging. Neurologia I Neurochirurgia Polska, 49(6), 349-353. https://doi.org/10.1016/j.pjnns.2015.09.003
Neurocognitive Disorders. (2013). En Diagnostic and Statistical Manual of Mental Disorders. American Psychiatric Association. https://doi.org/10.1176/appi.books.9780890425596.dsm17
Nuwer, M. R., Lehmann, D., da Silva, F. L., Matsuoka, S., Sutherling, W., & Vibert, J. F. (1999). IFCN guidelines for topographic and frequency analysis of EEGs and EPs.The International Federation of Clinical Neurophysiology. Electroencephalography and Clinical Neurophysiology. Supplement, 52, 15-20.
O’Driscoll, C., & Shaikh, M. (2017). Cross-Cultural Applicability of the Montreal Cognitive Assessment (MoCA): A Systematic Review. Journal of Alzheimer’s Disease: JAD, 58(3), 789-801. https://doi.org/10.3233/JAD-161042
Okamura, N., Harada, R., Furumoto, S., Arai, H., Yanai, K., & Kudo, Y. (2014). Tau PET Imaging in Alzheimer’s Disease. Current Neurology and Neuroscience Reports, 14(11). Scopus. https://doi.org/10.1007/s11910-014-0500-6
Olabarrieta-Landa, L., Rivera, D., Galarza-del-Angel, J., Garza, M., Saracho, C., Rodríguez, W., Chávez-Oliveros, M., Rábago, B., Leibach, G., Schebela, S., Martínez, C., Luna, M., Longoni, M., Ocampo-Barba, N., Rodríguez, G., Aliaga, A., Esenarro, L., García de la Cadena, C., Perrin, B., & Arango-Lasprilla, J. (2015). Verbal fluency tests: Normative data for the Latin American Spanish speaking adult population. NeuroRehabilitation, 37(4), 515-561. https://doi.org/10.3233/NRE-151279
OMS | La demencia: Una prioridad para la salud pública. (s. f.). WHO; World Health Organization. Recuperado 5 de diciembre de 2020, de http://www.who.int/mental_health/neurology/dementia/es/
Onishi, J., Suzuki, Y., Yoshiko, K., Hibino, S., & Iguchi, A. (2005). Predictive model for assessing cognitive impairment by quantitative electroencephalography. Cognitive and Behavioral Neurology: Official Journal of the Society for Behavioral and Cognitive Neurology, 18(3), 179-184. https://doi.org/10.1097/01.wnn.0000178227.54315.38
Oostenveld, R., Fries, P., Maris, E., & Schoffelen, J.-M. (2010). FieldTrip: Open Source Software for Advanced Analysis of MEG, EEG, and Invasive Electrophysiological Data. Computational Intelligence and Neuroscience, 2011, e156869. https://doi.org/10.1155/2011/156869
Oosterman, J. M., Oosterveld, S., Rikkert, M. G. O., Claassen, J. A., & Kessels, R. P. C. (2012). Medial temporal lobe atrophy relates to executive dysfunction in Alzheimer’s disease. International Psychogeriatrics, 24(9), 1474-1482. https://doi.org/10.1017/S1041610212000506
Oppenheim, A. V. (1999). Discrete-time signal processing. Pearson Education India.
Oppenheim, A. V., Buck, J. R., & Schafer, R. W. (2001). Discrete-time signal processing. Vol. 2. Upper Saddle River, NJ: Prentice Hall.
Orta-Salazar, E., Cuellar-Lemus, C. A., Díaz-Cintra, S., & Feria-Velasco, A. I. (2014). Marcaje colinérgico en la corteza cerebral y el hipocampo en algunas especies animales y su relación con la enfermedad de Alzheimer. Neurología, 29(8), 497-503. https://doi.org/10.1016/j.nrl.2012.10.007
Paddick, S.-M., Gray, W. K., McGuire, J., Richardson, J., Dotchin, C., & Walker, R. W. (2017). Cognitive screening tools for identification of dementia in illiterate and low educated older adults, a systematic reviewand meta-analysis. International Psychogeriatrics, 29(6), 897-929. Scopus. https://doi.org/10.1017/S1041610216001976
Pascual-Marqui, R. D. (2007). Discrete, 3D distributed, linear imaging methods of electric neuronal activity. Part 1: Exact, zero error localization. arXiv:0710.3341 [math-ph, physics:physics, q-bio]. http://arxiv.org/abs/0710.3341
Pasquier, F., Leys, D., Weerts, J. G., Mounier-Vehier, F., Barkhof, F., & Scheltens, P. (1996). Inter- and intraobserver reproducibility of cerebral atrophy assessment on MRI scans with hemispheric infarcts. European Neurology, 36(5), 268-272. https://doi.org/10.1159/000117270
Pavlov, I. P. (1927). Conditional reflexes: An investigation of the physiological activity of the cerebral cortex
Pavlov, I. P. (1941). Lectures on conditioned reflexes. Vol. II. Conditioned reflexes and psychiatry. http://psycnet.apa.org/psycinfo/1941-04246-000
Pavlov, I. P., & Thompson, W. H. (1902). The work of the digestive glands. Charles Griffin. http://scholar.google.com/scholar?cluster=18207307312015685009&hl=en&oi=schol arr
Pedraza, O. L., Salazar, A. M., Sierra, F. A., Soler, D., Castro, J., Castillo, P., Hernández, A., Piñeros, C., Pedraza, O. L., Salazar, A. M., Sierra, F. A., Soler, D., Castro, J., Castillo, P., Hernández, A., & Piñeros, C. (2016). Contabilidad, validez de criterio y discriminante del Montreal Cognitive Assessment (MoCA) test, en un grupo de adultos de Bogotá. Acta Medica Colombiana, 41(4), 221-228.
Pennanen, C., Kivipelto, M., Tuomainen, S., Hartikainen, P., Hänninen, T., Laakso, M. P., Hallikainen, M., Vanhanen, M., Nissinen, A., Helkala, E.-L., Vainio, P., Vanninen, R., Partanen, K., & Soininen, H. (2004). Hippocampus and entorhinal cortex in mild cognitive impairment and early AD. Neurobiology of Aging, 25(3), 303-310. https://doi.org/10.1016/S0197-4580(03)00084-8
Pereda, E., Quiroga, R. Q., & Bhattacharya, J. (2005). Nonlinear multivariate analysis of neurophysiological signals. Progress in Neurobiology, 77(1-2), 1-37. https://doi.org/10.1016/j.pneurobio.2005.10.003
Pessoa, R. M. P., Bomfim, A. J. L., Ferreira, B. L. C., & Chagas, M. H. N. (2019). Diagnostic criteria and prevalence of mild cognitive impairment in older adults living in the community: A systematic review and meta-analysis. Revista de Psiquiatria Clinica, 46(3), 72-79. Scopus. https://doi.org/10.1590/0101-60830000000197
Petersen, R. C., Lopez, O., Armstrong, M. J., Getchius, T. S. D., Ganguli, M., Gloss, D., Gronseth, G. S., Marson, D., Pringsheim, T., Day, G. S., Sager, M., Stevens, J., & Rae-Grant, A. (2018). Practice guideline update summary: Mild cognitive impairment: Report of the Guideline Development, Dissemination, and Implementation Subcommittee of the American Academy of Neurology. Neurology, 90(3), 126-135. https://doi.org/10.1212/WNL.0000000000004826
Petersen, R. C., Smith, G. E., Waring, S. C., Ivnik, R. J., Tangalos, E. G., & Kokmen, E. (1999). Mild cognitive impairment: Clinical characterization and outcome. Archives of Neurology, 56(3), 303-308. https://doi.org/10.1001/archneur.56.3.303
Petrella, J. R., Wang, L., Krishnan, S., Slavin, M. J., Prince, S. E., Tran, T.-T. T., & Doraiswamy, P. M. (2007). Cortical deactivation in mild cognitive impairment: High field-strength functional MR imaging. Radiology, 245(1), 224-235. https://doi.org/10.1148/radiol.2451061847
Pievani, M., Filippini, N., van den Heuvel, M. P., Cappa, S. F., & Frisoni, G. B. (2014). Brain connectivity in neurodegenerative diseases—From phenotype to proteinopathy. Nature Reviews. Neurology, 10(11), 620-633. https://doi.org/10.1038/nrneurol.2014.178
Pihlajamäki, M., Jauhiainen, A. M., & Soininen, H. (2009). Structural and functional MRI in mild cognitive impairment. Current Alzheimer Research, 6(2), 179-185. https://doi.org/10.2174/156720509787602898
Pijnenburg, Y. A. L., vd Made, Y., van Cappellen van Walsum, A. M., Knol, D. L., Scheltens, Ph., & Stam, C. J. (2004). EEG synchronization likelihood in mild cognitive impairment and Alzheimer’s disease during a working memory task. Clinical Neurophysiology, 115(6), 1332-1339. https://doi.org/10.1016/j.clinph.2003.12.029
Pikovsky, A., Rosenblum, M., & Kurths, J. (2003). Synchronization: A Universal Concept in Nonlinear Sciences.
Pinto, E., & Peters, R. (2009). Literature review of the Clock Drawing Test as a tool for cognitive screening. Dementia and geriatric cognitive disorders, 27(3), 201-213.
Pinto, T. C. C., Machado, L., Bulgacov, T. M., Rodrigues-Júnior, A. L., Costa, M. L. G., Ximenes, R. C. C., & Sougey, E. B. (2019). Is the Montreal Cognitive Assessment (MoCA) screening superior to the Mini-Mental State Examination (MMSE) in the detection of mild cognitive impairment (MCI) and Alzheimer’s Disease (AD) in the elderly? International Psychogeriatrics, 31(4), 491-504. Scopus. https://doi.org/10.1017/S1041610218001370
Poil, S.-S., de Haan, W., van der Flier, W. M., Mansvelder, H. D., Scheltens, P., & Linkenkaer-Hansen, K. (2013). Integrative EEG biomarkers predict progression to Alzheimer’s disease at the MCI stage. Frontiers in Aging Neuroscience, 5, 58. https://doi.org/10.3389/fnagi.2013.00058
Portelius, E., Hansson, S. F., Tran, A. J., Zetterberg, H., Grognet, P., Vanmechelen, E., Höglund, K., Brinkmalm, G., Westman-Brinkmalm, A., Nordhoff, E., Blennow, K., & Gobom, J. (2008). Characterization of tau in cerebrospinal fluid using mass spectrometry. Journal of Proteome Research, 7(5), 2114-2120. https://doi.org/10.1021/pr7008669
Poulin, S. P., Dautoff, R., Morris, J. C., Barrett, L. F., Dickerson, B. C., & Alzheimer’s Disease Neuroimaging Initiative. (2011). Amygdala atrophy is prominent in early Alzheimer’s disease and relates to symptom severity. Psychiatry Research, 194(1), 7-13. https://doi.org/10.1016/j.pscychresns.2011.06.014
Prichep, L. S. (2007). Quantitative EEG and Electromagnetic Brain Imaging in Aging and in the Evolution of Dementia. Annals of the New York Academy of Sciences, 1097(1), 156-167. https://doi.org/10.1196/annals.1379.008
Prichep, L. S., John, E. R., Ferris, S. H., Rausch, L., Fang, Z., Cancro, R., Torossian, C., & Reisberg, B. (2006). Prediction of longitudinal cognitive decline in normal elderly with subjective complaints using electrophysiological imaging. Neurobiology of Aging, 27(3), 471-481. https://doi.org/10.1016/j.neurobiolaging.2005.07.021
Prichep, L. S., John, E. R., Ferris, S. H., Reisberg, B., Almas, M., Alper, K., & Cancro, R. (1994). Quantitative EEG correlates of cognitive deterioration in the elderly. Neurobiology of Aging, 15(1), 85-90. https://doi.org/10.1016/0197-4580(94)90147-3
Primavera, A., Novello, P., Finocchi, C., Canevari, E., & Corsello, L. (1990). Correlation between Mini-Mental State examination and quantitative electroencephalography in senile dementia of Alzheimer type. Neuropsychobiology, 23(2), 74-78. Scopus. https://doi.org/10.1159/000119430
Puttaert, D., Coquelet, N., Wens, V., Peigneux, P., Fery, P., Rovai, A., Trotta, N., Sadeghi, N., Coolen, T., Bier, J.-C., Goldman, S., & De Tiège, X. (2020). Alterations in resting-state network dynamics along the Alzheimer’s disease continuum. Scientific Reports, 10(1), Art. 1. https://doi.org/10.1038/s41598-020-76201-3
Resting-state EEG source localization and functional connectivity in schizophrenia-like psychosis of epilepsy—PubMed. (s. f.). Recuperado 14 de marzo de 2022, de https://pubmed.ncbi.nlm.nih.gov/22125634/
Resumen-Ejecutivo-Encuesta-SABE.pdf. (s. f.). Recuperado 15 de marzo de 2022, de https://www.minsalud.gov.co/sites/rid/Lists/BibliotecaDigital/RIDE/VS/ED/GCFI/Resumen-Ejecutivo-Encuesta-SABE.pdf
Rivera, D., Ruiz, N. C., Gutiérrez-Hernández, C., & Calderón, J. A. (s. f.). Test de Aprendizaje Verbal de Hopkins-Revisado (HVLT-R). Datos normativos basados en regresiones múltiples para población colombiana. 13.
Roalf, D. R., Moberg, P. J., Xie, S. X., Wolk, D. A., Moelter, S. T., & Arnold, S. E. (2013). Comparative accuracies of two common screening instruments for the classification of Alzheimer’s disease, mild cognitive impairment and healthy aging. Alzheimer’s & dementia : the journal of the Alzheimer’s Association, 9(5), 529-537. https://doi.org/10.1016/j.jalz.2012.10.001
Rodrigues, S. M., Schafe, G. E., & LeDoux, J. E. (2001). Intra-Amygdala Blockade of the NR2B Subunit of the NMDA Receptor Disrupts the Acquisition But Not the Expression of Fear Conditioning. Journal of Neuroscience, 21(17), 6889-6896.
Rossini, P. M., Di Iorio, R., Vecchio, F., Anfossi, M., Babiloni, C., Bozzali, M., Bruni, A. C., Cappa, S. F., Escudero, J., Fraga, F. J., Giannakopoulos, P., Guntekin, B., Logroscino, G., Marra, C., Miraglia, F., Panza, F., Tecchio, F., Pascual-Leone, A., & Dubois, B. (2020). Early diagnosis of Alzheimer’s disease: The role of biomarkers including advanced EEG signal analysis. Report from the IFCN-sponsored panel of experts. Clinical Neurophysiology, 131(6), 1287-1310. https://doi.org/10.1016/j.clinph.2020.03.003
Salmon, D. P. (2008). Chapter 5 Neuropsychology of aging and dementia. Handbook of Clinical Neurology, 88, 113-135. Scopus. https://doi.org/10.1016/S0072-9752(07)88005-5
Samuraki, M., Matsunari, I., & Yamada, M. (2017). Neuroimaging study of Alzheimer’s disease in volunteer-based cohort. En Neuroimaging Diagnosis for Alzheimer’s Disease and Other Dementias (pp. 243-255). Scopus. https://doi.org/10.1007/978-4-431-55133-1_14
Sankari, Z., Adeli, H., & Adeli, A. (2012). Wavelet Coherence Model for Diagnosis of Alzheimer Disease. Clinical EEG and Neuroscience, 43(4), 268-278. https://doi.org/10.1177/1550059412444970
Scheltens, P., Leys, D., Barkhof, F., Huglo, D., Weinstein, H. C., Vermersch, P., Kuiper, M., Steinling, M., Wolters, E. C., & Valk, J. (1992). Atrophy of medial temporal lobes on MRI in «probable» Alzheimer’s disease and normal ageing: Diagnostic value and neuropsychological correlates. Journal of Neurology, Neurosurgery, and Psychiatry, 55(10), 967-972. https://doi.org/10.1136/jnnp.55.10.967
Schindler, S. E., Bollinger, J. G., Ovod, V., Mawuenyega, K. G., Li, Y., Gordon, B. A., Holtzman, D. M., Morris, J. C., Benzinger, T. L. S., Xiong, C., Fagan, A. M., & Bateman, R. J. (2019). High-precision plasma β-amyloid 42/40 predicts current and future brain amyloidosis. Neurology, 93(17), e1647-e1659. https://doi.org/10.1212/WNL.0000000000008081
Scoville, W. B., & Milner, B. (1957). Loss of recent memory after bilateral hippocampal lesions. Journal of neurology, neurosurgery, and psychiatry, 20(1), 11.
Sekeres, M. J., Moscovitch, M., & Winocur, G. (2017). Mechanisms of Memory Consolidation and Transformation. En Cognitive Neuroscience of Memory Consolidation (pp. 17-44). Springer.
Seppälä, T. T., Nerg, O., Koivisto, A. M., Rummukainen, J., Puli, L., Zetterberg, H., Pyykkö, O. T., Helisalmi, S., Alafuzoff, I., Hiltunen, M., Jääskeläinen, J. E., Rinne, J., Soininen, H., Leinonen, V., & Herukka, S. K. (2012). CSF biomarkers for Alzheimer disease correlate with cortical brain biopsy findings. Neurology, 78(20), 1568-1575. https://doi.org/10.1212/WNL.0b013e3182563bd0
Serra, L., Cercignani, M., Lenzi, D., Perri, R., Fadda, L., Caltagirone, C., Macaluso, E., & Bozzali, M. (2010). Grey and white matter changes at different stages of Alzheimer’s disease. Journal of Alzheimer’s Disease: JAD, 19(1), 147-159. https://doi.org/10.3233/JAD-2010-1223
Serra, L., Cercignani, M., Petrosini, L., Basile, B., Perri, R., Fadda, L., Spanò, B., Marra, C., Giubilei, F., Carlesimo, G. A., Caltagirone, C., & Bozzali, M. (2011). Neuroanatomical correlates of cognitive reserve in Alzheimer disease. Rejuvenation Research, 14(2), 143-151. https://doi.org/10.1089/rej.2010.1103
Shaughnessy, L., Sheard, S., Goldfarb, D., & Atri, A. (2019). Cognitive Assessment of Alzheimer’s Disease and Dementias in Clinical Practice: Pragmatics of Brief Instruments and Neuropsychological Evaluation. The Journal of Clinical Psychiatry, 80(4), MS18002BR2C. https://doi.org/10.4088/JCP.MS18002BR2C
Sherrington, C. S. (1910). Flexion-reflex of the limb, crossed extension-reflex, and reflex stepping and standing. The Journal of Physiology, 40(1-2), 28-121.
Shi, S.-H., Hayashi, Y., Petralia, R. S., Zaman, S. H., Wenthold, R. J., Svoboda, K., & Malinow, R. (1999). Rapid Spine Delivery and Redistribution of AMPA Receptors After Synaptic NMDA Receptor Activation. Science, 284(5421), 1811-1816. https://doi.org/10.1126/science.284.5421.1811
Shum, D. H., McFarland, K. A., & Bain, J. D. (1990). Construct validity of eight tests of attention: Comparison of normal and closed head injured samples. The Clinical Neuropsychologist, 4(2), 151-162.
Silva, A. J., Kogan, J. H., Frankland, P. W., & Kida, S. (1998). CREB and memory. Annual Review of Neuroscience, 21, 127-148. https://doi.org/10.1146/annurev.neuro.21.1.12
Sir Charles Sherrington—Nobel Lecture: Inhibition as a Coordinative Factor. (s. f.). Recuperado 11 de mayo de 2017, de https://www.nobelprize.org/nobel_prizes/medicine/laureates/1932/sherrington lecture.html
Small, S. A., Perera, G. M., DeLaPaz, R., Mayeux, R., & Stern, Y. (1999). Differential regional dysfunction of the hippocampal formation among elderly with memory decline and Alzheimer’s disease. Annals of Neurology, 45(4), 466-472. https://doi.org/10.1002/1531-8249(199904)45:4<466::aid-ana8>3.0.co;2-q
Smith, G. (2016). Dementia care: A practical approach. CRC Press. https://books.google.com/books?hl=en&lr=&id=6hRjDAAAQBAJ&oi=fnd&pg=PP1&dq=%22Ann+Hayes,+Lorraine+Shaw,+and+Grahame%22+%22Psychological+interventions+in%22+%22Parker+and+Robert+G.%22+%22Smith+and+Denise%22+%22Rylance+and+Donal%22+%22End+of+life%22+%22Knott+and+Denise%22+&ots=2g0s9zCAMk&sig=pE__72LTF6oiZoKm4x5-3CR3vos
Soltani Zangbar, H., Soltani Zangbar, H., Ghadiri, T., Seyedi Vafaee, M., Ebrahimi Kalan, A., Fallahi, S., Ghorbani, M., & Shahabi, P. (2020). Theta Oscillations through Hippocampal/Prefrontal Pathway: Importance in Cognitive Performances. Brain Connectivity, 10(4), 157-169. Scopus. https://doi.org/10.1089/brain.2019.0733
Sperling, R. A., Bates, J. F., Chua, E. F., Cocchiarella, A. J., Rentz, D. M., Rosen, B. R., Schacter, D. L., & Albert, M. S. (2003). FMRI studies of associative encoding in young and elderly controls and mild Alzheimer’s disease. Journal of Neurology, Neurosurgery, and Psychiatry, 74(1), 44-50. https://doi.org/10.1136/jnnp.74.1.44
Squire, L. R. (1992). Memory and the hippocampus: A synthesis from findings with rats, monkeys, and humans. Psychological Review, 99(2), 195-231.
Stam, C. J., van Cappellen van Walsum, A. M., Pijnenburg, Y. A. L., Berendse, H. W., de Munck, J. C., Scheltens, P., & van Dijk, B. W. (2002). Generalized Synchronization of MEG Recordings in Alzheimer’s Disease: Evidence for Involvement of the Gamma Band. Journal of Clinical Neurophysiology, 19(6), 562-574.
Stam, C. J., van der Made, Y., Pijnenburg, Y. a. L., & Scheltens, P. (2003). EEG synchronization in mild cognitive impairment and Alzheimer’s disease. Acta Neurologica Scandinavica, 108(2), 90-96. https://doi.org/10.1034/j.1600-0404.2003.02067.x
Stanton, P. K., & Sarvey, J. M. (1984). Blockade of long-term potentiation in rat hippocampal CA1 region by inhibitors of protein synthesis. Journal of Neuroscience, 4(12), 3080-3088.
Stefanowska, M. (1897). Les appendices terminaux des dendrites cérébraux et leurs différents états physiologiques. Hayez, Imprimeur de l’Académie royale des sciences, des lettres et des beaux-arts de Belgique.
Stomrud, E., Hansson, O., Minthon, L., Blennow, K., Rosén, I., & Londos, E. (2010). Slowing of EEG correlates with CSF biomarkers and reduced cognitive speed in elderly with normal cognition over 4 years. Neurobiology of Aging, 31(2), 215-223. https://doi.org/10.1016/j.neurobiolaging.2008.03.025
Strauss, E., Sherman, E. M., & Spreen, O. (2006). A compendium of neuropsychological tests: Administration, norms, and commentary. American chemical society.
Suárez-Calvet, M., Karikari, T. K., Ashton, N. J., Lantero Rodríguez, J., Milà-Alomà, M., Gispert, J. D., Salvadó, G., Minguillon, C., Fauria, K., Shekari, M., Grau-Rivera, O., Arenaza-Urquijo, E. M., Sala-Vila, A., Sánchez-Benavides, G., González-de Echávarri, J. M., Kollmorgen, G., Stoops, E., Vanmechelen, E., Zetterberg, H., … ALFA Study. (2020). Novel tau biomarkers phosphorylated at T181, T217 or T231 rise in the initial stages of the preclinical Alzheimer’s continuum when only subtle changes in Aβ pathology are detected. EMBO Molecular Medicine, 12(12), e12921. https://doi.org/10.15252/emmm.202012921
Tabaton, M., Nunzi, M. G., Xue, R., Usiak, M., Autilio-Gambetti, L., & Gambetti, P. (1994). Soluble amyloid beta-protein is a marker of Alzheimer amyloid in brain but not in cerebrospinal fluid. Biochemical and Biophysical Research Communications, 200(3), 1598-1603. https://doi.org/10.1006/bbrc.1994.1634
Tang, Y. P., Wang, H., Feng, R., Kyin, M., & Tsien, J. Z. (2001). Differential effects of enrichment on learning and memory function in NR2B transgenic mice. Neuropharmacology, 41(6), 779-790.
Tangalos, E. G., & Petersen, R. C. (2018). Mild Cognitive Impairment in Geriatrics. Clinics in Geriatric Medicine, 34(4), 563-589. Scopus. https://doi.org/10.1016/j.cger.2018.06.005
Tapiola, T., Alafuzoff, I., Herukka, S.-K., Parkkinen, L., Hartikainen, P., Soininen, H., & Pirttilä, T. (2009). Cerebrospinal fluid {beta}-amyloid 42 and tau proteins as biomarkers of Alzheimer-type pathologic changes in the brain. Archives of Neurology, 66(3), 382-389. https://doi.org/10.1001/archneurol.2008.596
Tatebe, H., Kasai, T., Ohmichi, T., Kishi, Y., Kakeya, T., Waragai, M., Kondo, M., Allsop, D., & Tokuda, T. (2017). Quantification of plasma phosphorylated tau to use as a biomarker for brain Alzheimer pathology: Pilot case-control studies including patients with Alzheimer’s disease and down syndrome. Molecular Neurodegeneration, 12(1), 63. https://doi.org/10.1186/s13024-017-0206-8
The Alzheimer’s Association QC program for CSF and blood biomarkers | University of Gothenburg. (s. f.). Recuperado 11 de marzo de 2022, de https://www.gu.se/en/neuroscience-physiology/the-alzheimers-association-qc program-for-csf-and-blood-biomarker
Thiyagesh, S. N., Farrow, T. F. D., Parks, R. W., Accosta-Mesa, H., Young, C., Wilkinson, I. D., Hunter, M. D., & Woodruff, P. W. R. (2009). The neural basis of visuospatial perception in Alzheimer’s disease and healthy elderly comparison subjects: An fMRI study. Psychiatry Research, 172(2), 109-116. https://doi.org/10.1016/j.pscychresns.2008.11.002
Tononi, G., Edelman, G. M., & Sporns, O. (1998). Complexity and coherency: Integrating information in the brain. Trends in Cognitive Sciences, 2(12), 474-484. https://doi.org/10.1016/S1364-6613(98)01259-5
Tononi, G., Sporns, O., & Edelman, G. M. (1994). A measure for brain complexity: Relating functional segregation and integration in the nervous system. Proceedings of the National Academy of Sciences of the United States of America, 91(11), 5033-5037. https://doi.org/10.1073/pnas.91.11.5033
Torisson, G., van Westen, D., Stavenow, L., Minthon, L., & Londos, E. (2015). Medial temporal lobe atrophy is underreported and may have important clinical correlates in medical inpatients. BMC Geriatrics, 15(1), 65. https://doi.org/10.1186/s12877-015-0066-4
Tse, D., Takeuchi, T., Kakeyama, M., Kajii, Y., Okuno, H., Tohyama, C., Bito, H., & Morris, R. G. (2011). Schema-dependent gene activation and memory encoding in neocortex. Science, 333(6044), 891-895.
Tulving, E. (1972). Episodic and semantic memory.
Tzourio-Mazoyer, N., Landeau, B., Papathanassiou, D., Crivello, F., Etard, O., Delcroix, N., Mazoyer, B., & Joliot, M. (2002). Automated Anatomical Labeling of Activations in SPM Using a Macroscopic Anatomical Parcellation of the MNI MRI Single-Subject Brain. NeuroImage, 15(1), 273-289.https://doi.org/10.1006/nimg.2001.0978
van den Heuvel, M. P., & Hulshoff Pol, H. E. (2010). Exploring the brain network: A review on resting-state fMRI functional connectivity. European Neuropsychopharmacology, 20(8), 519-534. Scopus. https://doi.org/10.1016/j.euroneuro.2010.03.008
van der Hiele, K., Vein, A. A., Reijntjes, R. H. a. M., Westendorp, R. G. J., Bollen, E. L. E. M., van Buchem, M. A., van Dijk, J. G., & Middelkoop, H. a. M. (2007). EEG correlates in the spectrum of cognitive decline. Clinical Neurophysiology: Official Journal of the International Federation of Clinical Neurophysiology, 118(9), 1931-1939. https://doi.org/10.1016/j.clinph.2007.05.070
van Kesteren, M. T., Fernández, G., Norris, D. G., & Hermans, E. J. (2010). Persistent schema-dependent hippocampal-neocortical connectivity during memory encoding and postencoding rest in humans. Proceedings of the National Academy of Sciences, 107(16), 7550-7555.
van Kesteren, M. T., Rijpkema, M., Ruiter, D. J., & Fernández, G. (2010). Retrieval of associative information congruent with prior knowledge is related to increased medial prefrontal activity and connectivity. Journal of Neuroscience, 30(47), 15888-15894.
VanderWeele, T. J. (2019). Principles of confounder selection. European Journal of Epidemiology, 34(3), 211-219. https://doi.org/10.1007/s10654-019-00494-6
Vanhoenacker, A.-S., Sneyers, B., De Keyzer, F., Heye, S., & Demaerel, P. (2017). Evaluation and clinical correlation of practical cut-offs for visual rating scales of atrophy: Normal aging versus mild cognitive impairment and Alzheimer’s disease. Acta Neurologica Belgica, 117(3), 661-669. Scopus. https://doi.org/10.1007/s13760-017-0777-8
Varela, F., Lachaux, J.-P., Rodriguez, E., & Martinerie, J. (2001). The brainweb: Phase synchronization and large-scale integration. Nature Reviews Neuroscience, 2(4), Art. 4. https://doi.org/10.1038/35067550
Vecchio, F., Miraglia, F., Iberite, F., Lacidogna, G., Guglielmi, V., Marra, C., Pasqualetti, P., Tiziano, F. D., & Rossini, P. M. (2018). Sustainable method for Alzheimer dementia prediction in mild cognitive impairment: Electroencephalographic connectivity and graph theory combined with apolipoprotein E: MCI Conversion. Annals of Neurology, 84(2), 302-314. https://doi.org/10.1002/ana.25289
Vecchio, F., Miraglia, F., Judica, E., Cotelli, M., Alù, F., & Rossini, P. M. (2020). Human brain networks: A graph theoretical analysis of cortical connectivity normative database from EEG data in healthy elderly subjects. GeroScience, 42(2), 575-584. https://doi.org/10.1007/s11357-020-00176-2
Vermunt, L., Sikkes, S. A. M., van den Hout, A., Handels, R., Bos, I., van der Flier, W. M., Kern, S., Ousset, P.-J., Maruff, P., Skoog, I., Verhey, F. R. J., Freund-Levi, Y., Tsolaki, M., Wallin, Å. K., Olde Rikkert, M., Soininen, H., Spiru, L., Zetterberg, H., Blennow, K., … ICTUS/DSA study groups. (2019). Duration of preclinical, prodromal, and dementia stages of Alzheimer’s disease in relation to age, sex, and APOE genotype. Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association, 15(7), 888-898. https://doi.org/10.1016/j.jalz.2019.04.001
Wahlund, L.-O., Westman, E., van Westen, D., Wallin, A., Shams, S., Cavallin, L., Larsson, E.-M., & From, the I. C. I. N. (ICINET). (2017). Imaging biomarkers of dementia: Recommended visual rating scales with teaching cases. Insights into Imaging, 8(1), 79-90. Scopus. https://doi.org/10.1007/s13244-016-0521-6
Wang, R., Wang, J., Yu, H., Wei, X., Yang, C., & Deng, B. (2015). Power spectral density and coherence analysis of Alzheimer’s EEG. Cognitive Neurodynamics, 9(3), 291-304. https://doi.org/10.1007/s11571-014-9325-x
Wanleenuwat, P., Iwanowski, P., & Kozubski, W. (2019). Alzheimer’s dementia: Pathogenesis and impact of cardiovascular risk factors on cognitive decline. Postgraduate Medicine, 131(7), 415-422. Scopus. https://doi.org/10.1080/00325481.2019.1657776
Welle (www.dw.com), D. (s. f.). UNESCO: Dos de cada diez personas en América Latina no tienen los niveles mínimos de compresión de lectura | DW | 06.09.2019. DW.COM. Recuperado 12 de marzo de 2022, de https://www.dw.com/es/unesco-dos-de-cada diez-personas-en-am%C3%A9rica-latina-no-tienen-los-niveles m%C3%ADnimos de-compresi%C3%B3n-de-lectura/a-50333467
Weltgesundheitsorganisation, & Alzheimer’s Disease International (Eds.). (2012). Dementia: A public health priority.
Willis, T. (1962). 1664. Cerebri anatome, cui accessit nervorum descriptio et usus. London: Martin & Allestry by University College London on, 5(09), 06.
Wysokiński, A., Zboralski, K., Orzechowska, A., Gałecki, P., Florkowski, A., & Talarowska, M. (2010). Normalization of the Verbal Fluency Test on the basis of results for healthy subjects, patients with schizophrenia, patients with organic lesions of the chronic nervous system and patients with type 1 and 2 diabetes. Archives of Medical Science, 6(3), 438-446.
Yetkin, F. Z., Rosenberg, R. N., Weiner, M. F., Purdy, P. D., & Cullum, C. M. (2006). FMRI of working memory in patients with mild cognitive impairment and probable Alzheimer’s disease. European Radiology, 16(1), 193-206. https://doi.org/10.1007/s00330-005-2794-x
Yi, H.-A., Won, K. S., Chang, H. W., & Kim, H. W. (2018). Association between white matter lesions and cerebral Aβ burden. PLoS ONE, 13(9). Scopus. https://doi.org/10.1371/journal.pone.0204313
Yin, Y., Edelman, G. M., & Vanderklish, P. W. (2002). The brain-derived neurotrophic factor enhances synthesis of Arc in synaptoneurosomes. Proceedings of the National Academy of Sciences, 99(4), 2368-2373. https://doi.org/10.1073/pnas.042693699
Yuste, R. (2010). Dendritic Spines. MIT Press.
Zegarra-Valdivia, J., Solís, L. D., & Chino-Vilca, B. (2019). Effectiveness of the photo-test front of the MMSE, for the screening of cognitive deterioration in Peruvian population. Revista Ecuatoriana de Neurologia, 28(1), 39-46. Scopus.
Zetterberg, H. (2016). Neurofilament Light: A Dynamic Cross-Disease Fluid Biomarker for Neurodegeneration. Neuron, 91(1), 1-3. https://doi.org/10.1016/j.neuron.2016.06.030
Zetterberg, H., Skillbäck, T., Mattsson, N., Trojanowski, J. Q., Portelius, E., Shaw, L. M., Weiner, M. W., Blennow, K., & Alzheimer’s Disease Neuroimaging Initiative. (2016). Association of Cerebrospinal Fluid Neurofilament Light Concentration With Alzheimer Disease Progression. JAMA Neurology, 73(1), 60-67. https://doi.org/10.1001/jamaneurol.2015.3037
Zhang, H., & Jacobs, J. (2015). Traveling theta waves in the human hippocampus. Journal of Neuroscience, 35(36), 12477-12487.
dc.rights.license.spa.fl_str_mv Atribución-NoComercial-CompartirIgual 4.0 Internacional (CC BY-NC-SA 4.0)
dc.rights.uri.spa.fl_str_mv https://creativecommons.org/licenses/by-nc-sa/4.0/
dc.rights.accessrights.spa.fl_str_mv info:eu-repo/semantics/closedAccess
dc.rights.coar.spa.fl_str_mv http://purl.org/coar/access_right/c_14cb
rights_invalid_str_mv Atribución-NoComercial-CompartirIgual 4.0 Internacional (CC BY-NC-SA 4.0)
https://creativecommons.org/licenses/by-nc-sa/4.0/
http://purl.org/coar/access_right/c_14cb
eu_rights_str_mv closedAccess
dc.format.extent.spa.fl_str_mv 185 páginas
dc.format.mimetype.spa.fl_str_mv application/pdf
dc.publisher.department.spa.fl_str_mv Ciencias de la Salud
dc.publisher.place.spa.fl_str_mv Barranquilla, Colombia
dc.publisher.program.spa.fl_str_mv Doctorado en Neurociencia Cognitiva Aplicada
institution Corporación Universidad de la Costa
bitstream.url.fl_str_mv https://repositorio.cuc.edu.co/bitstream/11323/10766/1/Perfil%20de%20conectividad%20cerebral%20y%20el%20rendimiento%20cognitivo%20en%20consulta.pdf
https://repositorio.cuc.edu.co/bitstream/11323/10766/2/license.txt
https://repositorio.cuc.edu.co/bitstream/11323/10766/3/Perfil%20de%20conectividad%20cerebral%20y%20el%20rendimiento%20cognitivo%20en%20consulta.pdf.txt
https://repositorio.cuc.edu.co/bitstream/11323/10766/4/Perfil%20de%20conectividad%20cerebral%20y%20el%20rendimiento%20cognitivo%20en%20consulta.pdf.jpg
bitstream.checksum.fl_str_mv f581aa4152a2081208cc2eb41bb9707d
2f9959eaf5b71fae44bbf9ec84150c7a
c4e7ee6ed4877b5f9fdb5b641c9554c0
68106b15834b5c5cd4a2253177585e76
bitstream.checksumAlgorithm.fl_str_mv MD5
MD5
MD5
MD5
repository.name.fl_str_mv Repositorio Universidad de La Costa
repository.mail.fl_str_mv bdigital@metabiblioteca.com
_version_ 1808400033548075008
spelling Atribución-NoComercial-CompartirIgual 4.0 Internacional (CC BY-NC-SA 4.0)https://creativecommons.org/licenses/by-nc-sa/4.0/info:eu-repo/semantics/closedAccesshttp://purl.org/coar/access_right/c_14cbAllegri, Ricardo Franciscoe54a8d9c0479ed60d7234722e91b37c7600Barceló, Ernesto6d2dd71e9e8d5b84f2fdd24eebf0ee3f600Cuesta, Pablo7e884c627f42893aea529b58cf5b3d64600Camargo Camargo, Loidac31962c3607f3f9cf5418604b65ca111Cochen, SilviaThomson, AlfredoFabian Román, Nestor2024-02-21T23:56:38Z2024-02-21T23:56:38Z2023https://hdl.handle.net/11323/10766Corporación Universidad de la CostaREDICUC-Repositorio.cuc.https://repositorio.cuc.edu.co/El abordaje diagnóstico de pacientes con enfermedades que comprometen la cognición es un reto para los sistemas de salud. El electroencefalograma es considerado una técnica útil para evaluar el funcionamiento cerebral, de bajo costo y accesible a la población general. El presente estudio indica que el análisis de la conectividad cerebral y su relación con las pruebas de tamización cognitiva pueden ser un adecuado biomarcador de la función cerebral tanto en las pacientes con enfermedad de Alzheimer como en la población general. Se realizaron análisis tanto en la distribución espectral como en la organización funcional. Se evidenció que el aumento de la potencia Theta se correlacionó negativamente con el desempeño obtenido en las pruebas de tamización cognitiva en toda la población. Por su parte, la conectividad funcional se vio afectada de manera heterogénea. Se demostró una disminución en la sincronización de las oscilaciones Gamma frontotemporales en pacientes con enfermedad de Alzheimer que se correlacionó negativamente con los resultados de las pruebas de tamización. Además, se encontró disminución en la sincronización Theta de similar localización que se correlacionó con la prueba de MOCA. Adicionalmente, se evidenció un aumento en la sincronización Alpha en toda la población que se correlacionó positivamente con los resultados de MOCA.The diagnostic approach to patients with diseases that compromise cognition is a challenge for health systems. The electroencephalogram is considered a useful technique to assess brain function, inexpensive and accessible to the general population. The present study indicates that the analysis of brain connectivity and its relationship with cognitive screening tests may be a suitable biomarker of brain function in both Alzheimer's disease patients and the general population. Analyses were performed on both spectral distribution and functional organization. It was shown that the increase in Theta power was negatively correlated with the performance obtained in the cognitive screening tests in the whole population. Functional connectivity was heterogeneously affected. A decrease in the synchronization of frontotemporal Gamma oscillations was demonstrated in patients with Alzheimer's disease that correlated negatively with the results of the screening tests. In addition, a decrease in Theta synchronization of similar localization was found that correlated with MOCA testing. Additionally, an increase in Alpha synchronization was evidenced in the entire population that correlated positively with MOCA results.Lista de tablas y figuras7 -- Introducción 9 -- Planteamiento del problema 11 -- Pregunta problema 17 – Objetivos 18 -- Objetivo General 18 -- Objetivo Específicos 18 – Justificación 19 -- Marco Teórico 22 -- Una breve revisión histórica de los antecedentes en el estudio de la memoria 22 -- De la memoria a las espinas dendríticas 24 -- Procesos de memoria 26 -- Conectividad neural 26 -- Potenciación a largo plazo 28 -- Bases moleculares de la potenciación a largo plazo 30 -- Consolidación de la memoria 33 -- Cambios celulares en la consolidación 35 -- Consolidación en los sistemas corticales 37 -- Envejecimiento y alteraciones de memoria 39 -- Marco conceptual 41 -- Deterioro cognitivo leve y demencia 41 -- Deterioro cognitivo leve (DCL) 42 -- Enfermedad de Alzheimer 45 -- Criterios diagnósticos de la enfermedad de Alzheimer 46 -- Biomarcadores en enfermedad de Alzheimer 47 -- Biomarcador en imágenes cerebrales 47 -- Biomarcadores en LCR. 53 -- Electroencefalograma como biomarcador de alteración cognitiva 57 -- Estado del arte 61 -- Estudios del EEG en DCL y DA 61 -- Metodología 64 -- Tipo y Diseño de Investigación 64 -- Población y muestra 64 -- Población 64 -- Muestra 64 -- Valoración clínica 64 -- Valoración paraclínica 64 -- Valoración neuropsicológica 65 -- Clasificación de los grupos 69 -- Criterios de Inclusión 69 -- Criterios de Exclusión 69 -- Técnicas e instrumentos 70 – Filtrado 72 -- Análisis del espectro de potencia 75 -- Análisis de sincronización: conectividad funcional 75 – Procedimiento 77 -- Consideraciones Éticas 78 -- Cuadro de Variables 78 – Resultados 80 -- Poder espectral: consideraciones metodológicas particulares 84 -- Resultados análisis de potencia 86 -- FOTOTEST 86 -- MOCA 90 -- Resultados análisis de conectividad funcional 93 -- Consideraciones metodológicas especiales 93 – FOTOTEST 94 – MOCA 98 – Discusión 111 -- Conectividad funcional 118 – Conclusiones 124 – Limitaciones 125 – Recomendaciones 126 -- Direcciones futuras 126 – Referencias 127 --Doctor(a) en Neurociencia Cognitiva AplicadaDoctorado185 páginasapplication/pdfspaPerfil de conectividad cerebral y el rendimiento cognitivo en consulta neurológicaTrabajo de grado - Doctoradohttp://purl.org/coar/resource_type/c_db06Textinfo:eu-repo/semantics/doctoralThesishttp://purl.org/redcol/resource_type/TDinfo:eu-repo/semantics/drafthttp://purl.org/coar/version/c_b1a7d7d4d402bcceCiencias de la SaludBarranquilla, ColombiaDoctorado en Neurociencia Cognitiva Aplicada2021 Alzheimer’s disease facts and figures. (2021). Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association, 17(3), 327-406. https://doi.org/10.1002/alz.12328280920-boletines-poblacionales-adulto-mayorI-2020.pdf. (s. f.). Recuperado 15 de marzo de 2022, de https://www.minsalud.gov.co/sites/rid/Lists/BibliotecaDigital/RIDE/DE/PS/280920- boletines-poblacionales-adulto-mayorI-2020.pdfAdler, G., Brassen, S., & Jajcevic, A. (2003). EEG coherence in Alzheimer’s dementia. Journal of Neural Transmission (Vienna, Austria: 1996), 110(9), 1051-1058. https://doi.org/10.1007/s00702-003-0024-8Adler, G., Brassen, S., & Jajcevic, A. (2003). EEG coherence in Alzheimer’s dementia. Journal of Neural Transmission (Vienna, Austria: 1996), 110(9), 1051-1058. https://doi.org/10.1007/s00702-003-0024-8Aggarwal, N. T. (2005). Mild cognitive impairment in different functional domains and incident Alzheimer’s disease. Journal of Neurology, Neurosurgery & Psychiatry, 76(11), 1479-1484. https://doi.org/10.1136/jnnp.2004.053561Ahmed, R. M., Paterson, R. W., Warren, J. D., Zetterberg, H., O’Brien, J. T., Fox, N. C., Halliday, G. M., & Schott, J. M. (2014). Biomarkers in dementia: Clinical utility and new directions. Journal of Neurology, Neurosurgery & Psychiatry, 85(12), 1426- 1434. https://doi.org/10.1136/jnnp-2014-307662Albert, M. S., DeKosky, S. T., Dickson, D., Dubois, B., Feldman, H. H., Fox, N. C., Gamst, A., Holtzman, D. M., Jagust, W. J., Petersen, R. C., Snyder, P. J., Carrillo, M. C., Thies, B., & Phelps, C. H. (2011a). The diagnosis of mild cognitive impairment due to Alzheimer’s disease: Recommendations from the National Institute on Aging Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s and Dementia, 7(3), 270-279. Scopus. https://doi.org/10.1016/j.jalz.2011.03.008Albert, M. S., DeKosky, S. T., Dickson, D., Dubois, B., Feldman, H. H., Fox, N. C., Gamst, A., Holtzman, D. M., Jagust, W. J., Petersen, R. C., Snyder, P. J., Carrillo, M. C., Thies, B., & Phelps, C. H. (2011b). The diagnosis of mild cognitive impairment due to Alzheimer’s disease: Recommendations from the National Institute on Aging Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s & Dementia, 7(3), 270-279. https://doi.org/10.1016/j.jalz.2011.03.008Al-Nuaimi, A. H. H., Jammeh, E., Sun, L., & Ifeachor, E. (2018). Complexity Measures for Quantifying Changes in Electroencephalogram in Alzheimer’s Disease. Complexity, 2018, e8915079. https://doi.org/10.1155/2018/8915079Al-Nuaimi, A. H., Jammeh, E., Sun, L., & Ifeachor, E. (2015). Tsallis entropy as a biomarker for detection of Alzheimer’s disease. 2015 37th Annual International Conference of the IEEE Engineering in Medicine and Biology Society (EMBC), 4166-4169.Al-nuaimi, A. H., Jammeh, E., Sun, L., & Ifeachor, E. (2017). Higuchi fractal dimension of the electroencephalogram as a biomarker for early detection of Alzheimer’s disease. 2017 39th Annual International Conference of the IEEE Engineering in Medicine and Biology Society (EMBC), 2320-2324. https://doi.org/10.1109/EMBC.2017.8037320Altomare, D., Caprioglio, C., Assal, F., Allali, G., Mendes, A., Ribaldi, F., Ceyzeriat, K., Martins, M., Tomczyk, S., Stampacchia, S., Dodich, A., Boccardi, M., Chicherio, C., Frisoni, G. B., & Garibotto, V. (2021). Diagnostic value of amyloid-PET and tau PET: A head-to-head comparison. European Journal of Nuclear Medicine and Molecular Imaging, 48(7), 2200-2211. Scopus. https://doi.org/10.1007/s00259-021- 05246-xAlzheimer’s Association. (2014). 2014 Alzheimer’s disease facts and figures. Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association, 10(2), e47-92.Andersson, E., Janelidze, S., Lampinen, B., Nilsson, M., Leuzy, A., Stomrud, E., Blennow, K., Zetterberg, H., & Hansson, O. (2020). Blood and cerebrospinal fluid neurofilament light differentially detect neurodegeneration in early Alzheimer’s disease. Neurobiology of Aging, 95, 143-153. https://doi.org/10.1016/j.neurobiolaging.2020.07.018Arango-Lasprilla, J. C., Rivera, D., Garza, M. T., Saracho, C. P., Rodríguez, W., Rodríguez Agudelo, Y., Aguayo, A., Schebela, S., Luna, M., Longoni, M., Martínez, C., Doyle, S., Ocampo-Barba, N., Galarza-del-Angel, J., Aliaga, A., Bringas, M., Esenarro, L., García-Egan, P., & Perrin, P. B. (2015). Hopkins Verbal Learning Test– Revised: Normative data for the Latin American Spanish speaking adult population. NeuroRehabilitation, 37(4), 699-718. https://doi.org/10.3233/NRE-151286Aron, L., & Yankner, B. A. (2016). Neural synchronization in Alzheimer’s disease. Nature, 540(7632), Art. 7632. https://doi.org/10.1038/540207aArráez-Aybar, L.-A., Navia-Álvarez, P., Fuentes-Redondo, T., & Bueno-López, J.-L. (2015). Thomas Willis, a pioneer in translational research in anatomy (on the 350th anniversary of Cerebri anatome). Journal of Anatomy, 226(3), 289-300. https://doi.org/10.1111/joa.12273Ashburner, J., & Friston, K. J. (2000). Voxel-based morphometry—The methods. NeuroImage, 11(6 Pt 1), 805-821. https://doi.org/10.1006/nimg.2000.0582Association, A. P. (2014). Guía de consulta de los criterios diagnósticos del DSM-5®: Spanish Edition of the Desk Reference to the Diagnostic Criteria From DSM-5®. American Psychiatric Pub.Ávila, G. M. R. (2007). Sincronizando con el caos. REBOLDES.Axmacher, N., & Rasch, B. (2017). Cognitive neuroscience of memory consolidation. Springer.Babiloni, C., Arakaki, X., Azami, H., Bennys, K., Blinowska, K., Bonanni, L., Bujan, A., Carrillo, M. C., Cichocki, A., de Frutos-Lucas, J., Del Percio, C., Dubois, B., Edelmayer, R., Egan, G., Epelbaum, S., Escudero, J., Evans, A., Farina, F., Fargo, K., … Guntekin, B. (2021). Measures of resting state EEG rhythms for clinical trials in Alzheimer’s disease: Recommendations of an expert panel. Alzheimer’s & Dementia, 17(9), 1528-1553. https://doi.org/10.1002/alz.12311Babiloni, C., Barry, R. J., Başar, E., Blinowska, K. J., Cichocki, A., Drinkenburg, W. H. I. M., Klimesch, W., Knight, R. T., Lopes da Silva, F., Nunez, P., Oostenveld, R., Jeong, J., Pascual-Marqui, R., Valdes-Sosa, P., & Hallett, M. (2020). International Federation of Clinical Neurophysiology (IFCN) – EEG research workgroup: Recommendations on frequency and topographic analysis of resting state EEG rhythms. Part 1: Applications in clinical research studies. Clinical Neurophysiology, 131(1), 285-307. https://doi.org/10.1016/j.clinph.2019.06.234Babiloni, C., Binetti, G., Cassarino, A., Dal Forno, G., Del Percio, C., Ferreri, F., Ferri, R., Frisoni, G., Galderisi, S., Hirata, K., Lanuzza, B., Miniussi, C., Mucci, A., Nobili, F., Rodriguez, G., Luca Romani, G., & Rossini, P. M. (2006). Sources of cortical rhythms in adults during physiological aging: A multicentric EEG study. Human Brain Mapping, 27(2), 162-172. https://doi.org/10.1002/hbm.20175Babiloni, C., Del Percio, C., Caroli, A., Salvatore, E., Nicolai, E., Marzano, N., Lizio, R., Cavedo, E., Landau, S., Chen, K., Jagust, W., Reiman, E., Tedeschi, G., Montella, P., De Stefano, M., Gesualdo, L., Frisoni, G. B., & Soricelli, A. (2016). Cortical sources of resting state EEG rhythms are related to brain hypometabolism in subjects with Alzheimer’s disease: An EEG-PET study. Neurobiology of Aging, 48, 122-134. https://doi.org/10.1016/j.neurobiolaging.2016.08.021Babiloni, C., Del Percio, C., Lizio, R., Noce, G., Cordone, S., Lopez, S., Soricelli, A., Ferri, R., Pascarelli, M. T., Nobili, F., Arnaldi, D., Famà, F., Aarsland, D., Orzi, F., Buttinelli, C., Giubilei, F., Onofrj, M., Stocchi, F., Stirpe, P., … De Pandis, M. F. (2017). Abnormalities of Cortical Neural Synchronization Mechanisms in Subjects with Mild Cognitive Impairment due to Alzheimer’s and Parkinson’s Diseases: An EEG Study. Journal of Alzheimer’s Disease, 59(1), 339-358. https://doi.org/10.3233/JAD-160883Babiloni, C., Del Percio, C., Lizio, R., Noce, G., Lopez, S., Soricelli, A., Ferri, R., Pascarelli, M. T., Catania, V., Nobili, F., Arnaldi, D., Famà, F., Orzi, F., Buttinelli, C., Giubilei, F., Bonanni, L., Franciotti, R., Onofrj, M., Stirpe, P., … Stocchi, F. (2018). Functional cortical source connectivity of resting state electroencephalographic alpha rhythms shows similar abnormalities in patients with mild cognitive impairment due to Alzheimer’s and Parkinson’s diseases. Clinical Neurophysiology, 129(4), 766-782. https://doi.org/10.1016/j.clinph.2018.01.009Babiloni, C., Ferri, R., Binetti, G., Vecchio, F., Frisoni, G. B., Lanuzza, B., Miniussi, C., Nobili, F., Rodriguez, G., Rundo, F., Cassarino, A., Infarinato, F., Cassetta, E., Salinari, S., Eusebi, F., & Rossini, P. M. (2009). Directionality of EEG synchronization in Alzheimer’s disease subjects. Neurobiology of Aging, 30(1), 93- 102. https://doi.org/10.1016/j.neurobiolaging.2007.05.007Babiloni, C., Frisoni, G. B., Pievani, M., Vecchio, F., Lizio, R., Buttiglione, M., Geroldi, C., Fracassi, C., Eusebi, F., Ferri, R., & Rossini, P. M. (2009). Hippocampal volume and cortical sources of EEG alpha rhythms in mild cognitive impairment and Alzheimer disease. NeuroImage, 44(1), 123-135. https://doi.org/10.1016/j.neuroimage.2008.08.005Bajo, R., Castellanos, N. P., Cuesta, P., Aurtenetxe, S., Garcia-Prieto, J., Gil-Gregorio, P., del-Pozo, F., & Maestu, F. (2012). Differential Patterns of Connectivity in Progressive Mild Cognitive Impairment. Brain Connectivity, 2(1), 21-24. https://doi.org/10.1089/brain.2011.0069Balderas, I., Ramírez-Amaya, V., & Bermúdez-Rattoni, F. (2004). [Memory-linked morphological changes]. Revista De Neurologia, 38(10), 944-948.Barco, A., Alarcon, J. M., & Kandel, E. R. (2002). Expression of constitutively active CREB protein facilitates the late phase of long-term potentiation by enhancing synaptic capture. Cell, 108(5), 689-703.Barco, A., & Marie, H. (2011). Genetic approaches to investigate the role of CREB in neuronal plasticity and memory. Molecular neurobiology, 44(3), 330-349.Bartus, R. T., Dean, R. L., Beer, B., & Lippa, A. S. (1982). The cholinergic hypothesis of geriatric memory dysfunction. Science (New York, N.Y.), 217(4558), 408-414. https://doi.org/10.1126/science.7046051Bateman, R. J., Xiong, C., Benzinger, T. L. S., Fagan, A. M., Goate, A., Fox, N. C., Marcus, D. S., Cairns, N. J., Xie, X., Blazey, T. M., Holtzman, D. M., Santacruz, A., Buckles, V., Oliver, A., Moulder, K., Aisen, P. S., Ghetti, B., Klunk, W. E., McDade, E., … Morris, J. C. (2012). Clinical and Biomarker Changes in Dominantly Inherited Alzheimer’s Disease. New England Journal of Medicine, 367(9), 795-804. https://doi.org/10.1056/NEJMoa1202753Bauer, R. M., Grande, L., & Valenstein, E. (2003). Amnesic disorders.Benussi, A., Cantoni, V., Grassi, M., Brechet, L., Michel, C. M., Datta, A., Thomas, C., Gazzina, S., Cotelli, M. S., Bianchi, M., Premi, E., Gadola, Y., Cotelli, M., Pengo, M., Perrone, F., Scolaro, M., Archetti, S., Solje, E., Padovani, A., … Borroni, B. (2022). Increasing Brain Gamma Activity Improves Episodic Memory and Restores Cholinergic Dysfunction in Alzheimer’s Disease. Annals of Neurology, 92(2), 322- 334. Scopus. https://doi.org/10.1002/ana.26411Besthorn, C., Sattel, H., Geiger-Kabisch, C., Zerfass, R., & Förstl, H. (1995). Parameters of EEG dimensional complexity in Alzheimer’s disease. Electroencephalography and Clinical Neurophysiology, 95(2), 84-89. https://doi.org/10.1016/0013-4694(95)00050- 9Bethus, I., Tse, D., & Morris, R. G. (2010). Dopamine and memory: Modulation of the persistence of memory for novel hippocampal NMDA receptor-dependent paired associates. Journal of Neuroscience, 30(5), 1610-1618.Biomarkers Definitions Working Group. (2001). Biomarkers and surrogate endpoints: Preferred definitions and conceptual framework. Clinical Pharmacology and Therapeutics, 69(3), 89-95. https://doi.org/10.1067/mcp.2001.113989Black, M. M., & Baas, P. W. (1989). The basis of polarity in neurons. Trends in Neurosciences, 12(6), 211-214. https://doi.org/10.1016/0166-2236(89)90124-0Blennow, K., Dubois, B., Fagan, A. M., Lewczuk, P., de Leon, M. J., & Hampel, H. (2015). Clinical utility of cerebrospinal fluid biomarkers in the diagnosis of early Alzheimer’s disease. Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association, 11(1), 58-69. https://doi.org/10.1016/j.jalz.2014.02.004Blennow, K., Vanmechelen, E., & Hampel, H. (2001). CSF total tau, Abeta42 and phosphorylated tau protein as biomarkers for Alzheimer’s disease. Molecular Neurobiology, 24(1-3), 87-97. https://doi.org/10.1385/MN:24:1-3:087Blennow, K., Wallin, A., Agren, H., Spenger, C., Siegfried, J., & Vanmechelen, E. (1995). Tau protein in cerebrospinal fluid: A biochemical marker for axonal degeneration in Alzheimer disease? Molecular and Chemical Neuropathology, 26(3), 231-245. https://doi.org/10.1007/BF02815140Blennow, K., & Zetterberg, H. (2015). Understanding biomarkers of neurodegeneration: Ultrasensitive detection techniques pave the way for mechanistic understanding. Nature Medicine, 21(3), 217-219. https://doi.org/10.1038/nm.3810Bliss, T. V., & Collingridge, G. L. (1993). A synaptic model of memory: Long-term potentiation in the hippocampus. Nature, 361(6407), 31-39. https://doi.org/10.1038/361031a0Bliss, T. V. P., & Cooke, S. F. (2011). Long-term potentiation and long-term depression: A clinical perspective. Clinics, 66(Suppl 1), 3-17. https://doi.org/10.1590/S1807- 59322011001300002Blom, E. S., Giedraitis, V., Zetterberg, H., Fukumoto, H., Blennow, K., Hyman, B. T., Irizarry, M. C., Wahlund, L.-O., Lannfelt, L., & Ingelsson, M. (2009). Rapid progression from mild cognitive impairment to Alzheimer’s disease in subjects with elevated levels of tau in cerebrospinal fluid and the APOE epsilon4/epsilon4 genotype. Dementia and Geriatric Cognitive Disorders, 27(5), 458-464. https://doi.org/10.1159/000216841Boada, M., Cejudo, J. C., Tàrraga, L., López, O. L., & Kaufer, D. (2002). Neuropsychiatric Inventory Questionnaire (NPI-Q): Spanish validation of a brief clinical form of the Neuropsychiatric Inventory (NPI). Neurologia, 17(6), 317-323. Scopus.Bozzali, M., Filippi, M., Magnani, G., Cercignani, M., Franceschi, M., Schiatti, E., Castiglioni, S., Mossini, R., Falautano, M., Scotti, G., Comi, G., & Falini, A. (2006). The contribution of voxel-based morphometry in staging patients with mild cognitive impairment. Neurology, 67(3), 453-460. https://doi.org/10.1212/01.wnl.0000228243.56665.c2Braak, H., & Braak, E. (1991). Neuropathological stageing of Alzheimer-related changes. Acta Neuropathologica, 82(4), 239-259. https://doi.org/10.1007/BF00308809Brandt, J. (1991). The Hopkins Verbal Learning Test: Development of a new memory test with six equivalent forms. The clinical neuropsychologist, 5(2), 125-142.Brandt, J., & Benedict, R. H. (2001). Hopkins verbal learning test—Revised: Professional manual. Psychological Assessment Resources.Brenner, R. P., Ulrich, R. F., Spiker, D. G., Sclabassi, R. J., Reynolds, C. F., Marin, R. S., & Boller, F. (1986). Computerized EEG spectral analysis in elderly normal, demented and depressed subjects. Electroencephalography and Clinical Neurophysiology, 64(6), 483-492. https://doi.org/10.1016/0013-4694(86)90184-7Brier, M. R., Thomas, J. B., Snyder, A. Z., Benzinger, T. L., Zhang, D., Raichle, M. E., Holtzman, D. M., Morris, J. C., & Ances, B. M. (2012). Loss of intranetwork and internetwork resting state functional connections with Alzheimer’s disease progression. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, 32(26), 8890-8899. https://doi.org/10.1523/JNEUROSCI.5698- 11.2012Buckner, R. L. (2012). The serendipitous discovery of the brain’s default network. NeuroImage, 62(2), 1137-1145. https://doi.org/10.1016/j.neuroimage.2011.10.035Busse, A., Angermeyer, M. C., & Riedel-Heller, S. G. (2006). Progression of mild cognitive impairment to dementia: A challenge to current thinking. The British Journal of Psychiatry: The Journal of Mental Science, 189, 399-404. https://doi.org/10.1192/bjp.bp.105.014779Buzsáki, G. (2002). Theta Oscillations in the Hippocampus. Neuron, 33(3), 325-340. https://doi.org/10.1016/S0896-6273(02)00586-XCabeza, R., & Nyberg, L. (2000). Imaging cognition II: An empirical review of 275 PET and fMRI studies. Journal of Cognitive Neuroscience, 12(1), 1-47. Scopus. https://doi.org/10.1162/08989290051137585Cacho, J., García-García, R., Arcaya, J., Vicente, J. L., & Lantada, N. (1999). Una propuesta de aplicación y puntuación del test del reloj en la enfermedad de Alzheimer. Rev Neurol, 28(7), 648-655.Cajal, S. R. (1890). Textura de las circunvolutiones cerebrales de los mamíferos inferiores: Nota preventiva.Cajal, S. R. Y. (1894). The Croonian Lecture: La Fine Structure des Centres Nerveux. Proceedings of the Royal Society of London, 55(331-335), 444-468. https://doi.org/10.1098/rspl.1894.0063Cajal, S. R. y. (1899). Textura del sistema nervioso del hombre y de los vertebrados: Estudios sobre el plan estructural y composición histológica de los centros nerviosos adicionados de consideraciones fisiológicas fundadas en los nuevos descubrimentos. N. Moya.Cajal, S. R. y. (2012). Texture of the Nervous System of Man and the Vertebrates. Springer Science & Business Media.Cajal, S. R. y, & Cajal, C. S. de I. C. (Spain) I. R. y. (1952). Neuronismo o reticularismo?: Las pruebas objetivas de la unidad anatomica de las celulas nerviosas. Edicion preparada por el Instituto Cajal con motivo del primer centenario de su fundador y maestro.Camozzato, A. L., Godinho, C., Kochhann, R., Massochini, G., & Chaves, M. L. (2015). Validity of the Brazilian version of the Neuropsychiatric Inventory Questionnaire (NPI-Q). Arquivos de Neuro-Psiquiatria, 73(1), 41-45. Scopus. https://doi.org/10.1590/0004-282X20140177Can, S. S., Gencay-Can, A., & Gunendi, Z. (2012). Validity and reliability of the clock drawing test as a screening tool for cognitive impairment in patients with fibromyalgia. Comprehensive psychiatry, 53(1), 81-86.Carnero Pardo, C. (2011). El FOTOTEST en el cribado de demencia en atención primaria. Revista de Neurología, 52(10), 639. https://doi.org/10.33588/rn.5210.2010808Carnero Pardo, C., Sáez-Zea, C., Montiel Navarro, L., Del Saz, P., Feria Vilar, I., Pérez Navarro, M. J., Ruiz-Giménez, J., Vilchez-Carrillo, R., & Montoro-Rios, M. T. (2007). Diagnostic accuracy of the Phototest for cognitive impairment and dementia. Neurologia, 22(10), 860-869. Scopus.Carnero-Pardo, C., Sáez-Zea, C., Montiel-Navarro, L., Feria-Vilar, I., & Gurpegui, M. (2011). Estudio normativo y de fiabilidad del FOTOTEST. Neurología, 26(1), 20-25. https://doi.org/10.1016/j.nrl.2010.09.021Casadio, A., Martin, K. C., Giustetto, M., Zhu, H., Chen, M., Bartsch, D., Bailey, C. H., & Kandel, E. R. (1999). A transient, neuron-wide form of CREB-mediated long-term facilitation can be stabilized at specific synapses by local protein synthesis. Cell, 99(2), 221-237.Cassani, R., Estarellas, M., San-Martin, R., Fraga, F. J., & Falk, T. H. (2018). Systematic Review on Resting-State EEG for Alzheimer’s Disease Diagnosis and Progression Assessment. Disease Markers, 2018, e5174815. https://doi.org/10.1155/2018/5174815Casula, E. P., Pellicciari, M. C., Bonnì, S., Borghi, I., Maiella, M., Assogna, M., Minei, M., Motta, C., D’Acunto, A., Porrazzini, F., Pezzopane, V., Mencarelli, L., Roncaioli, A., Rocchi, L., Spampinato, D. A., Caltagirone, C., Santarnecchi, E., Martorana, A., & Koch, G. (2022). Decreased Frontal Gamma Activity in Alzheimer Disease Patients. Annals of Neurology, 92(3), 464-475. Scopus. https://doi.org/10.1002/ana.26444Cavedo, E., Boccardi, M., Ganzola, R., Canu, E., Beltramello, A., Caltagirone, C., Thompson, P. M., & Frisoni, G. B. (2011). Local amygdala structural differences with 3T MRI in patients with Alzheimer disease. Neurology, 76(8), 727-733. https://doi.org/10.1212/WNL.0b013e31820d62d9Cecchetti, G., Agosta, F., Basaia, S., Cividini, C., Cursi, M., Santangelo, R., Caso, F., Minicucci, F., Magnani, G., & Filippi, M. (2021). Resting-state electroencephalographic biomarkers of Alzheimer’s disease. NeuroImage: Clinical, 31. Scopus. https://doi.org/10.1016/j.nicl.2021.102711Cerebri anatome: Cui accessit Nervorum descriptio & usus. (1676). apud Joannem Maximilianum Lucas.Chandra, A., Dervenoulas, G., Politis, M., & for, the A. D. N. I. (2019). Magnetic resonance imaging in Alzheimer’s disease and mild cognitive impairment. Journal of Neurology, 266(6), 1293-1302. Scopus. https://doi.org/10.1007/s00415-018-9016-3Chen, A. C., Feng, W., Zhao, H., Yin, Y., & Wang, P. (2008). EEG default mode network in the human brain: Spectral regional field powers. Neuroimage, 41(2), 561-574.Choi, J., Ku, B., You, Y. G., Jo, M., Kwon, M., Choi, Y., Jung, S., Ryu, S., Park, E., Go, H., Kim, G., Cha, W., & Kim, J. U. (2019). Resting-state prefrontal EEG biomarkers in correlation with MMSE scores in elderly individuals. Scientific Reports, 9(1). Scopus. https://doi.org/10.1038/s41598-019-46789-2Cieslak, A., Smith, E. E., Lysack, J., & Ismail, Z. (2017). Case series of mild behavioral impairment: Toward an understanding of the early stages of neurodegenerative diseases affecting behavior and cognition. International Psychogeriatrics, 1-8. https://doi.org/10.1017/S1041610217001855Clute-Reinig, N., Jayadev, S., Rhoads, K., & Le Ny, A.-L. (2021). Alzheimer’s Disease Diagnostics Must Be Globally Accessible. Journal of Alzheimer’s Disease: JAD, 84(4), 1453-1455. https://doi.org/10.3233/JAD-210663Constitución de Colombia. (1991). ConstitucionColombia.com. https://www.constitucioncolombia.com/titulo-2/capitulo-2/articulo-46Counter, S. A., Buchanan, L. H., & Ortega, F. (2009). Neurophysiologic and neurocognitive case profiles of andean patients with chronic environmental lead poisoning. Journal of Toxicology and Environmental Health - Part A: Current Issues, 72(19), 1150-1159. Scopus. https://doi.org/10.1080/15287390903091772Coutanche, M. N., & Thompson-Schill, S. L. (2015). Rapid consolidation of new knowledge in adulthood via fast mapping. Trends in cognitive sciences, 19(9), 486-488Cowansage, K. K., Shuman, T., Dillingham, B. C., Chang, A., Golshani, P., & Mayford, M. (2014). Direct reactivation of a coherent neocortical memory of context. Neuron, 84(2), 432-441.Cuesta, P., Ochoa-Urrea, M., Funke, M., Hasan, O., Zhu, P., Marcos, A., López, M. E., Schulz, P. E., Lhatoo, S., Pantazis, D., Mosher, J. C., & Maestu, F. (2022). Gamma band functional connectivity reduction in patients with amnestic mild cognitive impairment and epileptiform activity. Brain Communications, 4(2). Scopus. ¿https://doi.org/10.1093/braincomms/fcac012Cuesta Prieto, P. (2015). Perfiles biomagnéticos de la actividad en reposo cerebral en las fases preclínica y prodrómica de la Enfermedad de Alzheimer: Influencia del alelo APOE-ε4 y de la acumulación de β-amiloide [Http://purl.org/dc/dcmitype/Text, Universidad Complutense de Madrid]. https://dialnet.unirioja.es/servlet/tesis?codigo=100398Dauwels, J., Srinivasan, K., Ramasubba Reddy, M., Musha, T., Vialatte, F.-B., Latchoumane, C., Jeong, J., & Cichocki, A. (2011). Slowing and Loss of Complexity in Alzheimer’s EEG: Two Sides of the Same Coin? International Journal of Alzheimer&#x2019;s Disease, 2011, e539621. https://doi.org/10.4061/2011/539621Davachi, L., & DuBrow, S. (2015). How the hippocampus preserves order: The role of prediction and context. Trends in cognitive sciences, 19(2), 92-99.Davis, D. H. J., Creavin, S. T., Yip, J. L. Y., Noel-Storr, A. H., Brayne, C., & Cullum, S. (2021). Montreal Cognitive Assessment for the detection of dementia. Cochrane Database of Systematic Reviews, 2021(7). Scopus. https://doi.org/10.1002/14651858.CD010775.pub3Davis, H. P., & Squire, L. R. (1984). Protein synthesis and memory: A review. Psychological bulletin, 96(3), 518.De Carlos, J. A., & Borrell, J. (2007). A historical reflection of the contributions of Cajal and Golgi to the foundations of neuroscience. Brain Research Reviews, 55(1), 8-16. https://doi.org/10.1016/j.brainresrev.2007.03.010de Frutos-Lucas, J., Cuesta, P., Ramírez-Toraño, F., Nebreda, A., Cuadrado-Soto, E., Peral Suárez, Á., Lopez-Sanz, D., Bruña, R., Marcos-de Pedro, S., Delgado-Losada, M. L., López-Sobaler, A. M., Concepción Rodríguez-Rojo, I., Barabash, A., Serrano Rodriguez, J. M., Laws, S. M., Dolado, A. M., López-Higes, R., Brown, B. M., & Maestú, F. (2020). Age and APOE genotype affect the relationship between objectively measured physical activity and power in the alpha band, a marker of brain disease. Alzheimer’s Research & Therapy, 12, 113. https://doi.org/10.1186/s13195- 020-00681-8de Jong, L. W., van der Hiele, K., Veer, I. M., Houwing, J. J., Westendorp, R. G. J., Bollen, E. L. E. M., de Bruin, P. W., Middelkoop, H. A. M., van Buchem, M. A., & van der Grond, J. (2008). Strongly reduced volumes of putamen and thalamus in Alzheimer’s disease: An MRI study. Brain, 131(12), 3277-3285. https://doi.org/10.1093/brain/awn278De La Torre, J. C. (2012). Cardiovascular risk factors promote brain hypoperfusion leading to cognitive decline and dementia. Cardiovascular Psychiatry and Neurology. Scopus. https://doi.org/10.1155/2012/367516de Sánchez, C. R., Nariño, D., & Cerón, J. F. M. (2010). Epidemiología y carga de la Enfermedad de Alzheimer. Acta Neurol Colomb, 26(3), 1.DeFelipe, J. (2015a). The dendritic spine story: An intriguing process of discovery. Frontiers in Neuroanatomy, 9, 14. https://doi.org/10.3389/fnana.2015.00014DeFelipe, J. (2015b). The dendritic spine story: An intriguing process of discovery. Frontiers in Neuroanatomy, 9, 14. https://doi.org/10.3389/fnana.2015.00014Delbeuck, X., Van der Linden, M., & Collette, F. (2003). Alzheimer’ Disease as a Disconnection Syndrome? Neuropsychology Review, 13(2), 79-92. https://doi.org/10.1023/A:1023832305702Delgado Derio, C., Guerrero Bonnet, S., Troncoso Ponce, M., Araneda Yañez, A., Slachevsky Chonchol, A., & Behrens Pellegrino, M. I. (2013). Memoria, fluidez y orientación: Prueba de cribado de deterioro cognitivo en 5 minutos. Neurología, 28(7), 400-407. https://doi.org/10.1016/j.nrl.2012.10.001Demencia. (2020, septiembre 21). https://www.who.int/es/news-room/fact sheets/detail/dementiDevos, H., Gustafson, K., Liao, K., Ahmadnezhad, P., Estes, B., Martin, L. E., Mahnken, J. D., Brooks, W. M., & Burns, J. M. (2022). EEG/ERP evidence of possible hyperexcitability in older adults with elevated beta-amyloid. Translational Neurodegeneration, 11(1). Scopus. https://doi.org/10.1186/s40035-022-00282-5Díaz Cabezas, R., Marulanda Mejía, F., Arias, M., & Helena, M. (2013). Prevalence of cognitive impairment and dementia in people older 65 years in a Colombian urban population. Acta Neurológica Colombiana, 29(3), 141-151.Dierks, T., Jelic, V., Pascual-Marqui, R. D., Wahlund, L.-O., Julin, P., Linden, D. E. J., Maurer, K., Winblad, B., & Nordberg, A. (2000). Spatial pattern of cerebral glucose metabolism (PET) correlates with localization of intracerebral EEG-generators in Alzheimer’s disease. Clinical Neurophysiology, 111(10), 1817-1824. Scopus. https://doi.org/10.1016/S1388-2457(00)00427-2Dong, Y., Sharma, V. K., Chan, B. P.-L., Venketasubramanian, N., Teoh, H. L., Seet, R. C. S., Tanicala, S., Chan, Y. H., & Chen, C. (2010). The Montreal Cognitive Assessment (MoCA) is superior to the Mini-Mental State Examination (MMSE) for the detection of vascular cognitive impairment after acute stroke. Journal of the Neurological Sciences, 299(1-2), 15-18. https://doi.org/10.1016/j.jns.2010.08.051Dringenberg, H. C. (2000). Alzheimer’s disease: More than a ‘cholinergic disorder’ —evidence that cholinergic–monoaminergic interactions contribute to EEG slowing and dementia. Behavioural Brain Research, 115(2), 235-249. https://doi.org/10.1016/S0166-4328(00)00261-8Du, A. T., Schuff, N., Kramer, J. H., Ganzer, S., Zhu, X. P., Jagust, W. J., Miller, B. L., Reed, B. R., Mungas, D., Yaffe, K., Chui, H. C., & Weiner, M. W. (2004). Higher atrophy rate of entorhinal cortex than hippocampus in AD. Neurology, 62(3), 422-427. https://doi.org/10.1212/01.wnl.0000106462.72282.90Eichenbaum, H. (2011). The Cognitive Neuroscience of Memory: An Introduction. Oxford University Press, USA.Elwood, R. W. (1991). The Wechsler Memory Scale—Revised: Psychometric characteristics and clinical application. Neuropsychology review, 2(2), 179-201.Emsell, L., Vanhaute, H., Vansteelandt, K., De Winter, F.-L., Christiaens, D., Van den Stock, J., Vandenberghe, R., Van Laere, K., Sunaert, S., Bouckaert, F., & Vandenbulcke, M. (2022). An optimized MRI and PET based clinical protocol for improving the differential diagnosis of geriatric depression and Alzheimer’s disease. Psychiatry Research - Neuroimaging, 320. Scopus. https://doi.org/10.1016/j.pscychresns.2022.111443Envejecimiento y salud. (2022, octubre 1). https://www.who.int/es/news-room/fact sheets/detail/ageing-and-healtEstudio FOTOTRANS: estudio multicéntrico sobre la validez del FOTOTEST en condiciones de práctica clínica | Neurología. (s. f.). Recuperado 10 de marzo de 2022, de https://www.elsevier.es/es-revista-neurologia-295-articulo-estudio-fototrans-estudio multicentrico-sobre-S021348531100260XFazekas, F. (1989). Magnetic resonance signal abnormalities in asymptomatic individuals: Their incidence and functional correlates. European Neurology, 29(3), 164-168. https://doi.org/10.1159/000116401Fonseca, L. C., Tedrus, G. M., Prandi, L. R., Almeida, A. M., & Furlanetto, D. S. (2011). Alzheimer’s disease: Relationship between cognitive aspects and power and coherence EEG measures. Arquivos de Neuro-Psiquiatria, 69(6), 875-881. Scopus. https://doi.org/10.1590/s0004-282x2011000700005FOTOTEST online. (s. f.). Recuperado 25 de julio de 2022, de https://www.hipocampo.org/FOTOTESTOnline/FOTOTEST-enlinea.aspFox, C. A., & Barnard, J. W. (1957). A quantitative study of the Purkinje cell dendritic branchlets and their relationship to afferent fibres. Journal of Anatomy, 91(3), 299- 313.Frank, D. A., & Greenberg, M. E. (1994). CREB: a mediator of long-term memory from mollusks to mammals. Cell, 79(1), 5-8.Frey, U., & Morris, R. G. (1998). Synaptic tagging: Implications for late maintenance of hippocampal long-term potentiation. Trends in neurosciences, 21(5), 181-188.Friston, K. J. (1994). Functional and effective connectivity in neuroimaging: A synthesis. Human Brain Mapping, 2(1-2), 56-78. https://doi.org/10.1002/hbm.460020107Fuchs, M., Kastner, J., Wagner, M., Hawes, S., & Ebersole, J. S. (2002). A standardized boundary element method volume conductor model. Clinical Neurophysiology: Official Journal of the International Federation of Clinical Neurophysiology, 113(5), 702-712. https://doi.org/10.1016/s1388-2457(02)00030-5Gallistel, C. R., & Matzel, L. D. (2013). The neuroscience of learning: Beyond the Hebbian synapse. Annual Review of Psychology, 64, 169-200. https://doi.org/10.1146/annurev psych-113011-143807Ganguli, M., Dodge, H. H., Shen, C., & DeKosky, S. T. (2004). Mild cognitive impairment, amnestic type: An epidemiologic study. Neurology, 63(1), 115-121.García-Morales, P., Gich-Fullà, J., Guardia-Olmos, J., & Casanova, J. P. (1998). Series de dígitos, series automáticas y orientación: Normas ampliadas del Test Barcelona. Neurología (Barc., Ed. impr.), 13(S-6), 277-286.García-Sánchez, J. E., García, E., & Lucila Merino, M. (2010). Cien años de la bala mágica del Dr. Ehrlich (1909–2009). Enfermedades Infecciosas y Microbiología Clínica, 28(8), 521-533. https://doi.org/10.1016/j.eimc.2009.07.009Gasser, T., & Molinari, L. (1996). The analysis of the EEG. Statistical Methods in Medical Research, 5(1), 67-99. https://doi.org/10.1177/096228029600500105Ge, Q., Lin, Z.-C., Gao, Y.-X., & Zhang, J.-X. (2020). A Robust Discriminant Framework Based on Functional Biomarkers of EEG and Its Potential for Diagnosis of Alzheimer’s Disease. Healthcare, 8(4), Art. 4. https://doi.org/10.3390/healthcare8040476Gianotti, L. R. R., Künig, G., Lehmann, D., Faber, P. L., Pascual-Marqui, R. D., Kochi, K., & Schreiter-Gasser, U. (2007). Correlation between disease severity and brain electric LORETA tomography in Alzheimer’s disease. Clinical Neurophysiology: Official Journal of the International Federation of Clinical Neurophysiology, 118(1), 186- 196. https://doi.org/10.1016/j.clinph.2006.09.007Gili, T., Cercignani, M., Serra, L., Perri, R., Giove, F., Maraviglia, B., Caltagirone, C., & Bozzali, M. (2011). Regional brain atrophy and functional disconnection across Alzheimer’s disease evolution. Journal of Neurology, Neurosurgery, and Psychiatry, 82(1), 58-66. https://doi.org/10.1136/jnnp.2009.199935Golby, A., Silverberg, G., Race, E., Gabrieli, S., O’Shea, J., Knierim, K., Stebbins, G., & Gabrieli, J. (2005). Memory encoding in Alzheimer’s disease: An fMRI study of explicit and implicit memory. Brain: A Journal of Neurology, 128(Pt 4), 773-787. https://doi.org/10.1093/brain/awh40Gómez, F. R., Guerrero, C. E. D., Bernal, G. B., Pongutá, J. P. C., & Olarte, J. L. R. (s. f.). Politica colombiana de envejecimiento humano y vejez 2015—2024. 54.Gonzalez-Escamilla, G., Atienza, M., & Cantero, J. L. (2015). Impaired cortical oscillatory coupling in mild cognitive impairment: Anatomical substrate and ApoE4 effects. Brain Structure & Function, 220(3), 1721-1737. https://doi.org/10.1007/s00429-014-0757-1Gordon, B. A., Najmi, S., Hsu, P., Roe, C. M., Morris, J. C., & Benzinger, T. L. (2015). The effects of white matter hyperintensities and amyloid deposition on Alzheimer dementia. NeuroImage: Clinical, 8, 246-252.Gouveia, P. A. R., Brucki, S. M. D., Malheiros, S. M. F., & Bueno, O. F. A. (2007). Disorders in planning and strategy application in frontal lobe lesion patients. Brain and cognition, 63(3), 240-246.Gray, E. G. (1959). Electron Microscopy of Synaptic Contacts on Dendrite Spines of the Cerebral Cortex. Nature, 183(4675), 1592-1593. https://doi.org/10.1038/1831592a0Greicius, M. D., Krasnow, B., Reiss, A. L., & Menon, V. (2003). Functional connectivity in the resting brain: A network analysis of the default mode hypothesis. Proceedings of the National Academy of Sciences, 100(1), 253-258. https://doi.org/10.1073/pnas.0135058100Greicius, M. D., Srivastava, G., Reiss, A. L., & Menon, V. (2004). Default-mode network activity distinguishes Alzheimer’s disease from healthy aging: Evidence from functional MRI. Proceedings of the National Academy of Sciences of the United States of America, 101(13), 4637-4642. https://doi.org/10.1073/pnas.0308627101Gu, F., Meng, X., Shen, E., & Cai, Z. (2003). Can we measure consciousness with eeg complexities? International Journal of Bifurcation and Chaos, 13(03), 733-742. https://doi.org/10.1142/S0218127403006893Hall, B., Mak, E., Cervenka, S., Aigbirhio, F. I., Rowe, J. B., & O’Brien, J. T. (2017). In vivo tau PET imaging in dementia: Pathophysiology, radiotracer quantification, and a systematic review of clinical findings. Ageing Research Reviews, 36, 50-63. Scopus. https://doi.org/10.1016/j.arr.2017.03.002Handayani, N., Haryanto, F., Khotimah, S. N., Arif, I., & Taruno, W. P. (2018). Coherence and phase synchrony analyses of EEG signals in Mild Cognitive Impairment (MCI): A study of functional brain connectivity. Polish Journal of Medical Physics and Engineering, 24(1), 1-9. Scopus. https://doi.org/10.2478/pjmpe-2018-0001Hanslmayr, S., & Staudigl, T. (2014). How brain oscillations form memories—A processing based perspective on oscillatory subsequent memory effects. NeuroImage, 85, 648-655. https://doi.org/10.1016/j.neuroimage.2013.05.121Hebb, D. O. (1949). The organization of behavior: A neuropsychological approach. John Wiley & Sons.Hemmy, L. S., Linskens, E. J., Silverman, P. C., Miller, M. A., Talley, K. M. C., Taylor, B. C., Ouellette, J. M., Greer, N. L., Wilt, T. J., Butler, M., & Fink, H. A. (2020). Brief cognitive tests for distinguishing clinical Alzheimer-type dementia from mild cognitive impairment or normal cognition in older adults with suspected cognitive impairment: A systematic review. Annals of Internal Medicine, 172(10), 678-687. Scopus. https://doi.org/10.7326/M19-3889Henry, J. D., & Crawford, J. R. (2004). A meta-analytic review of verbal fluency performance in patients with traumatic brain injury. Neuropsychology, 18(4), 621.Horvath, A. (2018). EEG and ERP biomarkers of Alzheimer rsquo s disease a critical review. Frontiers in Bioscience, 23(1), 183-220. https://doi.org/10.2741/4587Hsu, Y.-H., Huang, S.-M., Lin, S.-Y., Yang, J.-J., Tu, M.-C., & Kuo, L.-W. (2022). Prospective Memory and Default Mode Network Functional Connectivity in Normal and Pathological Aging. Journal of Alzheimer’s Disease, 86(2), 753-762. Scopus. https://doi.org/10.3233/JAD-215293Huang, Y. Y., & Kandel, E. R. (1994). Recruitment of long-lasting and protein kinase A dependent long-term potentiation in the CA1 region of hippocampus requires repeated tetanization. Learning & Memory, 1(1), 74-82. https://doi.org/10.1101/lm.1.1.74Huey, E. D., Manly, J. J., Tang, M.-X., Schupf, N., Brickman, A. M., Manoochehri, M., Mez, J., DeCarli, C., Devanand, D. P., & Mayeux, R. (2013). Course and etiology of dysexecutive MCI in a community sample. Alzheimer’s & dementia : the journal of the Alzheimer’s Association, 9(6), 632-639. https://doi.org/10.1016/j.jalz.2012.10.014Hunt, K., Adamson, J., Hewitt, C., & Nazareth, I. (2011). Do women consult more than men? A review of gender and consultation for back pain and headache. Journal of Health Services Research and Policy, 16(2), 108-117. Scopus. https://doi.org/10.1258/jhsrp.2010.009131Huygens, C. (1673). Horologium Oscillatorium.Iaccarino, H. F., Singer, A. C., Martorell, A. J., Rudenko, A., Gao, F., Gillingham, T. Z., Mathys, H., Seo, J., Kritskiy, O., Abdurrob, F., Adaikkan, C., Canter, R. G., Rueda, R., Brown, E. N., Boyden, E. S., & Tsai, L.-H. (2016). Gamma frequency entrainment attenuates amyloid load and modifies microglia. Nature, 540(7632), Art. 7632. https://doi.org/10.1038/nature20587Ibanez, A., Parra, M. A., Butler, C., & Latin America and the Caribbean Consortium on Dementia (LAC-CD). (2021). The Latin America and the Caribbean Consortium on Dementia (LAC-CD): From Networking to Research to Implementation Science. Journal of Alzheimer’s Disease: JAD, 82(s1), S379-S394. https://doi.org/10.3233/JAD-201384Isaac, J. T., Nicoll, R. A., & Malenka, R. C. (1995). Evidence for silent synapses: Implications for the expression of LTP. Neuron, 15(2), 427-434.Ivan Pavlov—Biographical. (s. f.). Recuperado 4 de noviembre de 2017, de https://www.nobelprize.org/nobel_prizes/medicine/laureates/1904/pavlov-bio.htmlJack, C. R., Bennett, D. A., Blennow, K., Carrillo, M. C., Dunn, B., Haeberlein, S. B., Holtzman, D. M., Jagust, W., Jessen, F., Karlawish, J., Liu, E., Molinuevo, J. L., Montine, T., Phelps, C., Rankin, K. P., Rowe, C. C., Scheltens, P., Siemers, E., Snyder, H. M., … Contributors. (2018). NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association, 14(4), 535-562. https://doi.org/10.1016/j.jalz.2018.02.018Janelidze, S., Stomrud, E., Smith, R., Palmqvist, S., Mattsson, N., Airey, D. C., Proctor, N. K., Chai, X., Shcherbinin, S., Sims, J. R., Triana-Baltzer, G., Theunis, C., Slemmon, R., Mercken, M., Kolb, H., Dage, J. L., & Hansson, O. (2020). Cerebrospinal fluid p-tau217 performs better than p-tau181 as a biomarker of Alzheimer’s disease. Nature Communications, 11(1), 1683. https://doi.org/10.1038/s41467-020-15436-0Jarrett, J. T., Berger, E. P., & Lansbury, P. T. (1993). The carboxy terminus of the beta amyloid protein is critical for the seeding of amyloid formation: Implications for the pathogenesis of Alzheimer’s disease. Biochemistry, 32(18), 4693-4697. https://doi.org/10.1021/bi00069a001Jelic, V., Johansson, S.-E., Almkvist, O., Shigeta, M., Julin, P., Nordberg, A., Winblad, B., & Wahlund, L.-O. (2000). Quantitative electroencephalography in mild cognitive impairment: Longitudinal changes and possible prediction of Alzheimer’s disease. Neurobiology of Aging, 21(4), 533-540. https://doi.org/10.1016/S0197-4580(00)00153-6Jeong, J., Gore, J. C., & Peterson, B. S. (2001). Mutual information analysis of the EEG in patients with Alzheimer’s disease. Clinical Neurophysiology, 112(5), 827-835. https://doi.org/10.1016/S1388-2457(01)00513-2Johnson, N., Barion, A., Rademaker, A., Rehkemper, G., & Weintraub, S. (2004). The Activities of Daily Living Questionnaire: A validation study in patients with dementia. Alzheimer disease & associated disorders, 18(4), 223-230.Jongsiriyanyong, S., & Limpawattana, P. (2018). Mild Cognitive Impairment in Clinical Practice: A Review Article. American Journal of Alzheimer’s Disease and Other Dementias, 33(8), 500-507. Scopus. https://doi.org/10.1177/1533317518791401Josselyn, S. A., Köhler, S., & Frankland, P. W. (2015). Finding the engram. Nature Reviews Neuroscience, 16(9), 521-534. https://doi.org/10.1038/nrn4000Kandel, E. R. (2007). En busca de la memoria: El nacimiento de una nueva ciencia de la mente. Katz Editores.Kandel, E. R., Schwartz, J. H., & Jessell, T. M. (2000). Principios de neurociencia. McGraw Hill InteramericanaKandel, E. R., & Spencer, W. A. (1968). Cellular neurophysiological approaches in the study of learning. Physiological Reviews, 48(1), 65-134.Kapasi, A., DeCarli, C., & Schneider, J. A. (2017). Impact of multiple pathologies on the threshold for clinically overt dementia. Acta Neuropathologica, 134(2), 171-186. https://doi.org/10.1007/s00401-017-1717-7Karikari, T. K., Benedet, A. L., Ashton, N. J., Lantero Rodriguez, J., Snellman, A., Suárez Calvet, M., Saha-Chaudhuri, P., Lussier, F., Kvartsberg, H., Rial, A. M., Pascoal, T. A., Andreasson, U., Schöll, M., Weiner, M. W., Rosa-Neto, P., Trojanowski, J. Q., Shaw, L. M., Blennow, K., Zetterberg, H., & Alzheimer’s Disease Neuroimaging Initiative. (2021). Diagnostic performance and prediction of clinical progression of plasma phospho-tau181 in the Alzheimer’s Disease Neuroimaging Initiative. Molecular Psychiatry, 26(2), 429-442. https://doi.org/10.1038/s41380-020-00923-zKarikari, T. K., Pascoal, T. A., Ashton, N. J., Janelidze, S., Benedet, A. L., Rodriguez, J. L., Chamoun, M., Savard, M., Kang, M. S., Therriault, J., Schöll, M., Massarweh, G., Soucy, J.-P., Höglund, K., Brinkmalm, G., Mattsson, N., Palmqvist, S., Gauthier, S., Stomrud, E., … Blennow, K. (2020). Blood phosphorylated tau 181 as a biomarker for Alzheimer’s disease: A diagnostic performance and prediction modelling study using data from four prospective cohorts. The Lancet. Neurology, 19(5), 422-433. https://doi.org/10.1016/S1474-4422(20)30071-5Kauer, J. A., Malenka, R. C., & Nicoll, R. A. (1988). NMDA application potentiates synaptic transmission in the hippocampus. Nature, 334(6179), 250-252. https://doi.org/10.1038/334250a0Kichigina, V. F. (2019). Disturbances of theta and gamma oscillations in the brains with pathologies pecular to Alzheimer’s disease and temporal lobe epilepsy. Zhurnal Vysshei Nervnoi Deyatelnosti Imeni I.P. Pavlova, 69(4), 395-412. Scopus. https://doi.org/10.1134/S0044467719040075Klyucherev, T. O., Olszewski, P., Shalimova, A. A., Chubarev, V. N., Tarasov, V. V., Attwood, M. M., Syvänen, S., & Schiöth, H. B. (2022). Advances in the development of new biomarkers for Alzheimer’s disease. Translational Neurodegeneration, 11(1). Scopus. https://doi.org/10.1186/s40035-022-00296-zKoenig, T., Prichep, L., Dierks, T., Hubl, D., Wahlund, L. O., John, E. R., & Jelic, V. (2005). Decreased EEG synchronization in Alzheimer’s disease and mild cognitive impairment. Neurobiology of Aging, 26(2), 165-171. https://doi.org/10.1016/j.neurobiolaging.2004.03.008Kölliker, A., & Kölliker, A. von. (1863). Handbuch der Gewebelehre des Menschen: Für Aerzte und Studirende. Wilhelm Engelmann.Kowalski, J. W., Gawel, M., Pfeffer, A., & Barcikowska, M. (2001). The Diagnostic Value of EEG in Alzheimer Disease: Correlation With the Severity of Mental Impairment. Journal of Clinical Neurophysiology, 18(6), 570-575.Kulkarni, N., & Bairagi, V. (2018). Chapter Two—Electroencephalogram and Its Use in Clinical Neuroscience. En N. Kulkarni & V. Bairagi (Eds.), EEG-Based Diagnosis of Alzheimer Disease (pp. 25-35). Academic Press. https://doi.org/10.1016/B978-0-12-815392-5.00002-2Kwak, Y. T. (2006). Quantitative EEG findings in different stages of Alzheimer’s disease. Journal of Clinical Neurophysiology: Official Publication of the American Electroencephalographic Society, 23(5), 456-461. https://doi.org/10.1097/01.wnp.0000223453.47663.63Lamar, M., Boots, E. A., Arfanakis, K., Barnes, L. L., & Schneider, J. A. (2020). Common Brain Structural Alterations Associated with Cardiovascular Disease Risk Factors and Alzheimer’s Dementia: Future Directions and Implications. Neuropsychology Review, 30(4), 546-557. Scopus. https://doi.org/10.1007/s11065-020-09460-6Larson, J., & Munkácsy, E. (2015). Theta-Burst LTP. Brain research, 1621, 38-50. https://doi.org/10.1016/j.brainres.2014.10.034Lashley, K. S. (1950). In search of the engram.Laufs, H. (2008). Endogenous brain oscillations and related networks detected by surface EEG‐combined fMRI. Human brain mapping, 29(7), 762-769.Lee, S.-H., Park, Y.-M., Kim, D.-W., & Im, C.-H. (2010). Global synchronization index as a biological correlate of cognitive decline in Alzheimer’s disease. Neuroscience Research, 66(4), 333-339. Scopus. https://doi.org/10.1016/j.neures.2009.12.004Lempel, A., & Ziv, J. (1976). On the Complexity of Finite Sequences. IEEE Transactions on Information Theory, 22(1), 75-81. https://doi.org/10.1109/TIT.1976.1055501Lenzi, D., Serra, L., Perri, R., Pantano, P., Lenzi, G. L., Paulesu, E., Caltagirone, C., Bozzali, M., & Macaluso, E. (2011). Single domain amnestic MCI: A multiple cognitive domains fMRI investigation. Neurobiology of Aging, 32(9), 1542-1557. https://doi.org/10.1016/j.neurobiolaging.2009.09.006Leuzy, A., Cullen, N. C., Mattsson-Carlgren, N., & Hansson, O. (2021). Current advances in plasma and cerebrospinal fluid biomarkers in Alzheimer’s disease. Current Opinion in Neurology, 34(2), 266-274. https://doi.org/10.1097/WCO.0000000000000904Ley 1850 de 2017—Medidas de protección al adulto mayor en Colombia. (2018, mayo 10). Así Vamos en Salud - indicadores en salud normatividad derechos. https://www.asivamosensalud.org/politicas-publicas/normatividad-leyes/promocion social/ley-1850-de-2017-medidas-de-proteccion-alLi, C., Zheng, J., Wang, J., Gui, L., & Li, C. (2009). An fMRI stroop task study of prefrontal cortical function in normal aging, mild cognitive impairment, and Alzheimer’s disease. Current Alzheimer Research, 6(6), 525-530. https://doi.org/10.2174/156720509790147142Lizio, R., Del Percio, C., Marzano, N., Soricelli, A., Yener, G. G., Başar, E., Mundi, C., De Rosa, S., Triggiani, A. I., Ferri, R., Arnaldi, D., Nobili, F. M., Cordone, S., Lopez, S., Carducci, F., Santi, G., Gesualdo, L., Rossini, P. M., Cavedo, E., … Babiloni, C. (2016). Neurophysiological assessment of Alzheimer’s disease individuals by a single electroencephalographic marker. Journal of Alzheimer’s Disease: JAD, 49(1), 159-177. https://doi.org/10.3233/JAD-143042Lizio, R., Vecchio, F., Frisoni, G. B., Ferri, R., Rodriguez, G., & Babiloni, C. (2011). Electroencephalographic rhythms in Alzheimer’s disease. International Journal of Alzheimer’s Disease, 2011, 927573. https://doi.org/10.4061/2011/927573Lleó, A., Alcolea, D., Martínez-Lage, P., Scheltens, P., Parnetti, L., Poirier, J., Simonsen, A. H., Verbeek, M. M., Rosa-Neto, P., Slot, R. E. R., Tainta, M., Izaguirre, A., Reijs, B. L. R., Farotti, L., Tsolaki, M., Vandenbergue, R., Freund-Levi, Y., Verhey, F. R. J., Clarimón, J., … Blennow, K. (2019). Longitudinal cerebrospinal fluid biomarker trajectories along the Alzheimer’s disease continuum in the BIOMARKAPD study. Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association, 15(6), 742-753. https://doi.org/10.1016/j.jalz.2019.01.015Locatelli, T., Cursi, M., Liberati, D., Franceschi, M., & Comi, G. (1998). EEG coherence in Alzheimer’s disease. Electroencephalography and Clinical Neurophysiology, 106(3), 229-237. https://doi.org/10.1016/S0013-4694(97)00129-6Lomo, T. (1966). Frequency potentiation of excitatory synaptic activity in dentate area of hippocampal formation. Acta Physiologica Scandinavica, 128-.Lomo, T. (1971). Patterns of activation in a monosynaptic cortical pathway: The perforant path input to the dentate area of the hippocampal formation. Experimental Brain Research, 12(1), 18-45.Lømo, T. (2003). The discovery of long-term potentiation. Philosophical Transactions of the Royal Society B: Biological Sciences, 358(1432), 617-620. https://doi.org/10.1098/rstb.2002.1226López, N., Allegri, R., & Soto-Añari, M. (2014). Capacidad diagnóstica y validación preliminar del Test del Reloj, versión de Cacho a la Orden, para enfermedad de Alzheimer de grado leve en población chilena. Revista Ecuatoriana de Neurología, 23(1-3), 18-23.Loureiro, C. C. de S., García, C., Adana, L., Yacelga, T., Rodríguez Lorenzana, A., & Maruta, C. (2018). Uso del test de evaluación cognitiva de Montreal (MoCA) en América Latina: Revisión sistemática. Revista de Neurología, 66(12), 397. https://doi.org/10.33588/rn.6612.2017508Lu, W., Man, H., Ju, W., Trimble, W. S., MacDonald, J. F., & Wang, Y. T. (2001). Activation of synaptic NMDA receptors induces membrane insertion of new AMPA receptors and LTP in cultured hippocampal neurons. Neuron, 29(1), 243-254.Lüscher, C., & Frerking, M. (2001). Restless AMPA receptors: Implications for synaptic transmission and plasticity. Trends in Neurosciences, 24(11), 665-670Lynch, M. A. (2004). Long-term potentiation and memory. Physiological Reviews, 84(1), 87-136. https://doi.org/10.1152/physrev.00014.2003Maestú, F., Cuesta, P., Hasan, O., Fernandéz, A., Funke, M., & Schulz, P. E. (2019). The Importance of the Validation of M/Eeg with Current Biomarkers in Alzheimer’s Disease. Frontiers in Human Neuroscience, 13. https://www.frontiersin.org/article/10.3389/fnhum.2019.00017Malenka, R. C., Kauer, J. A., Zucker, R. S., & Nicoll, R. A. (1988). Postsynaptic calcium is sufficient for potentiation of hippocampal synaptic transmission. Science, 242(4875), 81-84. https://doi.org/10.1126/science.2845577Malinow, R., & Malenka, R. C. (2002). AMPA receptor trafficking and synaptic plasticity. Annual Review of Neuroscience, 25, 103-126. https://doi.org/10.1146/annurev.neuro.25.112701.142758Mallo, S. C., Patten, S. B., Ismail, Z., Pereiro, A. X., Facal, D., Otero, C., & Juncos-Rabadán, O. (2020). Does the neuropsychiatric inventory predict progression from mild cognitive impairment to dementia? A systematic review and meta-analysis. Ageing Research Reviews, 58. Scopus. https://doi.org/10.1016/j.arr.2019.101004Manning, J. R., Polyn, S. M., Baltuch, G. H., Litt, B., & Kahana, M. J. (2011). Oscillatory patterns in temporal lobe reveal context reinstatement during memory search. Proceedings of the National Academy of Sciences, 108(31), 12893-12897.Matsunari, I. (2012). MRI and PET in Alzheimer’s disease. Neurological Surgery, 40(12), 1051-1063. Scopus.Mattia, D., Babiloni, F., Romigi, A., Cincotti, F., Bianchi, L., Sperli, F., Placidi, F., Bozzao, A., Giacomini, P., Floris, R., & Marciani, M. G. (2003). Quantitative EEG and dynamic susceptibility contrast MRI in Alzheimer’s disease: A correlative study. Clinical Neurophysiology, 114(7), 1210-1216. Scopus. https://doi.org/10.1016/S1388-2457(03)00085-3Mazziotta, J. C., Toga, A. W., Evans, A., Fox, P., & Lancaster, J. (1995). A probabilistic atlas of the human brain: Theory and rationale for its development. The International Consortium for Brain Mapping (ICBM). NeuroImage, 2(2), 89-101. https://doi.org/10.1006/nimg.1995.1012McGeown, W. J., Shanks, M. F., Forbes-McKay, K. E., & Venneri, A. (2009). Patterns of brain activity during a semantic task differentiate normal aging from early Alzheimer’s disease. Psychiatry Research, 173(3), 218-227. https://doi.org/10.1016/j.pscychresns.2008.10.005McKhann, G. M., Knopman, D. S., Chertkow, H., Hyman, B. T., Jack, C. R., Kawas, C. H., Klunk, W. E., Koroshetz, W. J., Manly, J. J., Mayeux, R., Mohs, R. C., Morris, J. C., Rossor, M. N., Scheltens, P., Carrillo, M. C., Thies, B., Weintraub, S., & Phelps, C. H. (2011). The diagnosis of dementia due to Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s & Dementia, 7(3), 263-269. https://doi.org/10.1016/j.jalz.2011.03.005Miledi, R., & Slater, C. R. (1966). The action of calcium on neuronal synapses in the squid. The Journal of Physiology, 184(2), 473-498. https://doi.org/10.1113/jphysiol.1966.sp007927MINISTERIO DE SALUD Y PROTECCIÓN SOCIAL. (2013). Envejecimiento demografico-Colombia-1951-2020. OFICINA DE PROMOCIÓN SOCIAL. https://www.minsalud.gov.co/sites/rid/Lists/BibliotecaDigital/RIDE/DE/PS/Envejecimiento-demografico-Colombia-1951-2020.pdMöller, C., Van Der Flier, W. M., Versteeg, A., Benedictus, M. R., Wattjes, M. P., Koedam, E. L. G. M., Scheltens, P., Barkhof, F., & Vrenken, H. (2014). Quantitative regional validation of the visual rating scale for posterior cortical atrophy. European Radiology, 24(2), 397-404. Scopus. https://doi.org/10.1007/s00330-013-3025-5Monllor, P., Cervera-Ferri, A., Lloret, M.-A., Esteve, D., Lopez, B., Leon, J.-L., & Lloret, A. (2021). Electroencephalography as a non-invasive biomarker of alzheimer’s disease: A forgotten candidate to substitute CSF molecules? International Journal of Molecular Sciences, 22(19). Scopus. https://doi.org/10.3390/ijms221910889Montez, T., Linkenkaer-Hansen, K., van Dijk, B. W., & Stam, C. J. (2006). Synchronization likelihood with explicit time-frequency priors. NeuroImage, 33(4), 1117-1125. https://doi.org/10.1016/j.neuroimage.2006.06.066Moretti, D. V. (2015). Theta and alpha eeg frequency interplay in subjects with mild cognitive impairment: Evidence from EEG, MRI and spect brain modifications. Frontiers in Aging Neuroscience, 7(FEB). Scopus. https://doi.org/10.3389/fnagi.2015.00031Morris, J. C. (2006). Mild Cognitive Impairment Is Early-Stage Alzheimer Disease: Time to Revise Diagnostic Criteria. Archives of Neurology, 63(1), 15-16. https://doi.org/10.1001/archneur.63.1.15Morris, R. G., Anderson, E., Lynch, G. S., & Baudry, M. (1986). Selective impairment of learning and blockade of long-term potentiation by an N-methyl-D-aspartate receptor antagonist, AP5. Nature, 319(6056), 774-776. https://doi.org/10.1038/319774a0Morris, R. G. M. (2006). Elements of a neurobiological theory of hippocampal function: The role of synaptic plasticity, synaptic tagging and schemas. European Journal of Neuroscience, 23(11), 2829-2846. https://doi.org/10.1111/j.1460-9568.2006.04888.xMorris, R. G. M. (2013). NMDA receptors and memory encoding. Neuropharmacology, 74, 32-40. https://doi.org/10.1016/j.neuropharm.2013.04.014Mullany, P., & Lynch, M. A. (1997). Changes in protein synthesis and synthesis of the synaptic vesicle protein, synaptophysin, in entorhinal cortex following induction of long-term potentiation in dentate gyrus: An age-related study in the rat. Neuropharmacology, 36(7), 973-980.Muñoz-Neira, C., López, O. L., Riveros, R., Núñez-Huasaf, J., Flores, P., & Slachevsky, A. (2012). The Technology – Activities of Daily Living Questionnaire: A Version with a Technology-Related Subscale. Dementia and Geriatric Cognitive Disorders, 33(6), 361-371. https://doi.org/10.1159/000338606Murphy, D. D., & Segal, M. (1997). Morphological plasticity of dendritic spines in central neurons is mediated by activation of cAMP response element binding protein. Proceedings of the National Academy of Sciences, 94(4), 1482-1487.Musa, G., Henríquez, F., Muñoz-Neira, C., Delgado, C., Lillo, P., & Slachevsky, A. (2017). Utility of the Neuropsychiatric Inventory Questionnaire (NPI-Q) in the assessment of a sample of patients with Alzheimer’s disease in Chile. Dementia & Neuropsychologia, 11(2), 129-136. https://doi.org/10.1590/1980-57642016dn11- 020005Musaeus, C. S., Engedal, K., Høgh, P., Jelic, V., Mørup, M., Naik, M., Oeksengaard, A.-R., Snaedal, J., Wahlund, L.-O., Waldemar, G., & Andersen, B. B. (2018). EEG Theta Power Is an Early Marker of Cognitive Decline in Dementia due to Alzheimer’s Disease. Journal of Alzheimer’s Disease, 64(4), 1359-1371. https://doi.org/10.3233/JAD-180300Musaeus, C. S., Engedal, K., Høgh, P., Jelic, V., Mørup, M., Naik, M., Oeksengaard, A.-R., Snaedal, J., Wahlund, L.-O., Waldemar, G., & Andersen, B. B. (2019). Oscillatory connectivity as a diagnostic marker of dementia due to Alzheimer’s disease. Clinical Neurophysiology, 130(10), 1889-1899.https://doi.org/10.1016/j.clinph.2019.07.016Nakamura, A., Cuesta, P., Fernández, A., Arahata, Y., Iwata, K., Kuratsubo, I., Bundo, M., Hattori, H., Sakurai, T., Fukuda, K., Washimi, Y., Endo, H., Takeda, A., Diers, K., Bajo, R., Maestú, F., Ito, K., & Kato, T. (2018). Electromagnetic signatures of the preclinical and prodromal stages of Alzheimer’s disease. Brain, 141(5), 1470-1485. https://doi.org/10.1093/brain/awy044Nakamura, A., Kaneko, N., Villemagne, V. L., Kato, T., Doecke, J., Doré, V., Fowler, C., Li, Q.-X., Martins, R., Rowe, C., Tomita, T., Matsuzaki, K., Ishii, K., Ishii, K., Arahata, Y., Iwamoto, S., Ito, K., Tanaka, K., Masters, C. L., & Yanagisawa, K. (2018). High performance plasma amyloid-β biomarkers for Alzheimer’s disease. Nature, 554(7691), 249-254. https://doi.org/10.1038/nature25456Nasreddine, Z. S., Phillips, N. A., Bédirian, V., Charbonneau, S., Whitehead, V., Collin, I., Cummings, J. L., & Chertkow, H. (2005). The Montreal Cognitive Assessment, MoCA: A Brief Screening Tool For Mild Cognitive Impairment. Journal of the American Geriatrics Society, 53(4), 695-699. https://doi.org/10.1111/j.1532- 5415.2005.53221.xNatham, R., Amirthalingam, P., & Arunachalam, G. (2018). Comparison of Montreal Cognitive Assessment (MOCA) with Mini Mental State Examination (MMSE) on association between homocysteine and cognitive status in epilepsy patients with phenytoin monotherapy. Asia Pacific Journal of Counselling and Psychotherapy, 9(2), 160-170. https://doi.org/10.1080/21507686.2018.1483411Nesteruk, M., Nesteruk, T., Styczyńska, M., Barczak, A., Mandecka, M., Walecki, J., & Barcikowska-Kotowicz, M. (2015). Predicting the conversion of mild cognitive impairment to Alzheimer’s disease based on the volumetric measurements of the selected brain structures in magnetic resonance imaging. Neurologia I Neurochirurgia Polska, 49(6), 349-353. https://doi.org/10.1016/j.pjnns.2015.09.003Neurocognitive Disorders. (2013). En Diagnostic and Statistical Manual of Mental Disorders. American Psychiatric Association. https://doi.org/10.1176/appi.books.9780890425596.dsm17Nuwer, M. R., Lehmann, D., da Silva, F. L., Matsuoka, S., Sutherling, W., & Vibert, J. F. (1999). IFCN guidelines for topographic and frequency analysis of EEGs and EPs.The International Federation of Clinical Neurophysiology. Electroencephalography and Clinical Neurophysiology. Supplement, 52, 15-20.O’Driscoll, C., & Shaikh, M. (2017). Cross-Cultural Applicability of the Montreal Cognitive Assessment (MoCA): A Systematic Review. Journal of Alzheimer’s Disease: JAD, 58(3), 789-801. https://doi.org/10.3233/JAD-161042Okamura, N., Harada, R., Furumoto, S., Arai, H., Yanai, K., & Kudo, Y. (2014). Tau PET Imaging in Alzheimer’s Disease. Current Neurology and Neuroscience Reports, 14(11). Scopus. https://doi.org/10.1007/s11910-014-0500-6Olabarrieta-Landa, L., Rivera, D., Galarza-del-Angel, J., Garza, M., Saracho, C., Rodríguez, W., Chávez-Oliveros, M., Rábago, B., Leibach, G., Schebela, S., Martínez, C., Luna, M., Longoni, M., Ocampo-Barba, N., Rodríguez, G., Aliaga, A., Esenarro, L., García de la Cadena, C., Perrin, B., & Arango-Lasprilla, J. (2015). Verbal fluency tests: Normative data for the Latin American Spanish speaking adult population. NeuroRehabilitation, 37(4), 515-561. https://doi.org/10.3233/NRE-151279OMS | La demencia: Una prioridad para la salud pública. (s. f.). WHO; World Health Organization. Recuperado 5 de diciembre de 2020, de http://www.who.int/mental_health/neurology/dementia/es/Onishi, J., Suzuki, Y., Yoshiko, K., Hibino, S., & Iguchi, A. (2005). Predictive model for assessing cognitive impairment by quantitative electroencephalography. Cognitive and Behavioral Neurology: Official Journal of the Society for Behavioral and Cognitive Neurology, 18(3), 179-184. https://doi.org/10.1097/01.wnn.0000178227.54315.38Oostenveld, R., Fries, P., Maris, E., & Schoffelen, J.-M. (2010). FieldTrip: Open Source Software for Advanced Analysis of MEG, EEG, and Invasive Electrophysiological Data. Computational Intelligence and Neuroscience, 2011, e156869. https://doi.org/10.1155/2011/156869Oosterman, J. M., Oosterveld, S., Rikkert, M. G. O., Claassen, J. A., & Kessels, R. P. C. (2012). Medial temporal lobe atrophy relates to executive dysfunction in Alzheimer’s disease. International Psychogeriatrics, 24(9), 1474-1482. https://doi.org/10.1017/S1041610212000506Oppenheim, A. V. (1999). Discrete-time signal processing. Pearson Education India.Oppenheim, A. V., Buck, J. R., & Schafer, R. W. (2001). Discrete-time signal processing. Vol. 2. Upper Saddle River, NJ: Prentice Hall.Orta-Salazar, E., Cuellar-Lemus, C. A., Díaz-Cintra, S., & Feria-Velasco, A. I. (2014). Marcaje colinérgico en la corteza cerebral y el hipocampo en algunas especies animales y su relación con la enfermedad de Alzheimer. Neurología, 29(8), 497-503. https://doi.org/10.1016/j.nrl.2012.10.007Paddick, S.-M., Gray, W. K., McGuire, J., Richardson, J., Dotchin, C., & Walker, R. W. (2017). Cognitive screening tools for identification of dementia in illiterate and low educated older adults, a systematic reviewand meta-analysis. International Psychogeriatrics, 29(6), 897-929. Scopus. https://doi.org/10.1017/S1041610216001976Pascual-Marqui, R. D. (2007). Discrete, 3D distributed, linear imaging methods of electric neuronal activity. Part 1: Exact, zero error localization. arXiv:0710.3341 [math-ph, physics:physics, q-bio]. http://arxiv.org/abs/0710.3341Pasquier, F., Leys, D., Weerts, J. G., Mounier-Vehier, F., Barkhof, F., & Scheltens, P. (1996). Inter- and intraobserver reproducibility of cerebral atrophy assessment on MRI scans with hemispheric infarcts. European Neurology, 36(5), 268-272. https://doi.org/10.1159/000117270Pavlov, I. P. (1927). Conditional reflexes: An investigation of the physiological activity of the cerebral cortexPavlov, I. P. (1941). Lectures on conditioned reflexes. Vol. II. Conditioned reflexes and psychiatry. http://psycnet.apa.org/psycinfo/1941-04246-000Pavlov, I. P., & Thompson, W. H. (1902). The work of the digestive glands. Charles Griffin. http://scholar.google.com/scholar?cluster=18207307312015685009&hl=en&oi=schol arrPedraza, O. L., Salazar, A. M., Sierra, F. A., Soler, D., Castro, J., Castillo, P., Hernández, A., Piñeros, C., Pedraza, O. L., Salazar, A. M., Sierra, F. A., Soler, D., Castro, J., Castillo, P., Hernández, A., & Piñeros, C. (2016). Contabilidad, validez de criterio y discriminante del Montreal Cognitive Assessment (MoCA) test, en un grupo de adultos de Bogotá. Acta Medica Colombiana, 41(4), 221-228.Pennanen, C., Kivipelto, M., Tuomainen, S., Hartikainen, P., Hänninen, T., Laakso, M. P., Hallikainen, M., Vanhanen, M., Nissinen, A., Helkala, E.-L., Vainio, P., Vanninen, R., Partanen, K., & Soininen, H. (2004). Hippocampus and entorhinal cortex in mild cognitive impairment and early AD. Neurobiology of Aging, 25(3), 303-310. https://doi.org/10.1016/S0197-4580(03)00084-8Pereda, E., Quiroga, R. Q., & Bhattacharya, J. (2005). Nonlinear multivariate analysis of neurophysiological signals. Progress in Neurobiology, 77(1-2), 1-37. https://doi.org/10.1016/j.pneurobio.2005.10.003Pessoa, R. M. P., Bomfim, A. J. L., Ferreira, B. L. C., & Chagas, M. H. N. (2019). Diagnostic criteria and prevalence of mild cognitive impairment in older adults living in the community: A systematic review and meta-analysis. Revista de Psiquiatria Clinica, 46(3), 72-79. Scopus. https://doi.org/10.1590/0101-60830000000197Petersen, R. C., Lopez, O., Armstrong, M. J., Getchius, T. S. D., Ganguli, M., Gloss, D., Gronseth, G. S., Marson, D., Pringsheim, T., Day, G. S., Sager, M., Stevens, J., & Rae-Grant, A. (2018). Practice guideline update summary: Mild cognitive impairment: Report of the Guideline Development, Dissemination, and Implementation Subcommittee of the American Academy of Neurology. Neurology, 90(3), 126-135. https://doi.org/10.1212/WNL.0000000000004826Petersen, R. C., Smith, G. E., Waring, S. C., Ivnik, R. J., Tangalos, E. G., & Kokmen, E. (1999). Mild cognitive impairment: Clinical characterization and outcome. Archives of Neurology, 56(3), 303-308. https://doi.org/10.1001/archneur.56.3.303Petrella, J. R., Wang, L., Krishnan, S., Slavin, M. J., Prince, S. E., Tran, T.-T. T., & Doraiswamy, P. M. (2007). Cortical deactivation in mild cognitive impairment: High field-strength functional MR imaging. Radiology, 245(1), 224-235. https://doi.org/10.1148/radiol.2451061847Pievani, M., Filippini, N., van den Heuvel, M. P., Cappa, S. F., & Frisoni, G. B. (2014). Brain connectivity in neurodegenerative diseases—From phenotype to proteinopathy. Nature Reviews. Neurology, 10(11), 620-633. https://doi.org/10.1038/nrneurol.2014.178Pihlajamäki, M., Jauhiainen, A. M., & Soininen, H. (2009). Structural and functional MRI in mild cognitive impairment. Current Alzheimer Research, 6(2), 179-185. https://doi.org/10.2174/156720509787602898Pijnenburg, Y. A. L., vd Made, Y., van Cappellen van Walsum, A. M., Knol, D. L., Scheltens, Ph., & Stam, C. J. (2004). EEG synchronization likelihood in mild cognitive impairment and Alzheimer’s disease during a working memory task. Clinical Neurophysiology, 115(6), 1332-1339. https://doi.org/10.1016/j.clinph.2003.12.029Pikovsky, A., Rosenblum, M., & Kurths, J. (2003). Synchronization: A Universal Concept in Nonlinear Sciences.Pinto, E., & Peters, R. (2009). Literature review of the Clock Drawing Test as a tool for cognitive screening. Dementia and geriatric cognitive disorders, 27(3), 201-213.Pinto, T. C. C., Machado, L., Bulgacov, T. M., Rodrigues-Júnior, A. L., Costa, M. L. G., Ximenes, R. C. C., & Sougey, E. B. (2019). Is the Montreal Cognitive Assessment (MoCA) screening superior to the Mini-Mental State Examination (MMSE) in the detection of mild cognitive impairment (MCI) and Alzheimer’s Disease (AD) in the elderly? International Psychogeriatrics, 31(4), 491-504. Scopus. https://doi.org/10.1017/S1041610218001370Poil, S.-S., de Haan, W., van der Flier, W. M., Mansvelder, H. D., Scheltens, P., & Linkenkaer-Hansen, K. (2013). Integrative EEG biomarkers predict progression to Alzheimer’s disease at the MCI stage. Frontiers in Aging Neuroscience, 5, 58. https://doi.org/10.3389/fnagi.2013.00058Portelius, E., Hansson, S. F., Tran, A. J., Zetterberg, H., Grognet, P., Vanmechelen, E., Höglund, K., Brinkmalm, G., Westman-Brinkmalm, A., Nordhoff, E., Blennow, K., & Gobom, J. (2008). Characterization of tau in cerebrospinal fluid using mass spectrometry. Journal of Proteome Research, 7(5), 2114-2120. https://doi.org/10.1021/pr7008669Poulin, S. P., Dautoff, R., Morris, J. C., Barrett, L. F., Dickerson, B. C., & Alzheimer’s Disease Neuroimaging Initiative. (2011). Amygdala atrophy is prominent in early Alzheimer’s disease and relates to symptom severity. Psychiatry Research, 194(1), 7-13. https://doi.org/10.1016/j.pscychresns.2011.06.014Prichep, L. S. (2007). Quantitative EEG and Electromagnetic Brain Imaging in Aging and in the Evolution of Dementia. Annals of the New York Academy of Sciences, 1097(1), 156-167. https://doi.org/10.1196/annals.1379.008Prichep, L. S., John, E. R., Ferris, S. H., Rausch, L., Fang, Z., Cancro, R., Torossian, C., & Reisberg, B. (2006). Prediction of longitudinal cognitive decline in normal elderly with subjective complaints using electrophysiological imaging. Neurobiology of Aging, 27(3), 471-481. https://doi.org/10.1016/j.neurobiolaging.2005.07.021Prichep, L. S., John, E. R., Ferris, S. H., Reisberg, B., Almas, M., Alper, K., & Cancro, R. (1994). Quantitative EEG correlates of cognitive deterioration in the elderly. Neurobiology of Aging, 15(1), 85-90. https://doi.org/10.1016/0197-4580(94)90147-3Primavera, A., Novello, P., Finocchi, C., Canevari, E., & Corsello, L. (1990). Correlation between Mini-Mental State examination and quantitative electroencephalography in senile dementia of Alzheimer type. Neuropsychobiology, 23(2), 74-78. Scopus. https://doi.org/10.1159/000119430Puttaert, D., Coquelet, N., Wens, V., Peigneux, P., Fery, P., Rovai, A., Trotta, N., Sadeghi, N., Coolen, T., Bier, J.-C., Goldman, S., & De Tiège, X. (2020). Alterations in resting-state network dynamics along the Alzheimer’s disease continuum. Scientific Reports, 10(1), Art. 1. https://doi.org/10.1038/s41598-020-76201-3Resting-state EEG source localization and functional connectivity in schizophrenia-like psychosis of epilepsy—PubMed. (s. f.). Recuperado 14 de marzo de 2022, de https://pubmed.ncbi.nlm.nih.gov/22125634/Resumen-Ejecutivo-Encuesta-SABE.pdf. (s. f.). Recuperado 15 de marzo de 2022, de https://www.minsalud.gov.co/sites/rid/Lists/BibliotecaDigital/RIDE/VS/ED/GCFI/Resumen-Ejecutivo-Encuesta-SABE.pdfRivera, D., Ruiz, N. C., Gutiérrez-Hernández, C., & Calderón, J. A. (s. f.). Test de Aprendizaje Verbal de Hopkins-Revisado (HVLT-R). Datos normativos basados en regresiones múltiples para población colombiana. 13.Roalf, D. R., Moberg, P. J., Xie, S. X., Wolk, D. A., Moelter, S. T., & Arnold, S. E. (2013). Comparative accuracies of two common screening instruments for the classification of Alzheimer’s disease, mild cognitive impairment and healthy aging. Alzheimer’s & dementia : the journal of the Alzheimer’s Association, 9(5), 529-537. https://doi.org/10.1016/j.jalz.2012.10.001Rodrigues, S. M., Schafe, G. E., & LeDoux, J. E. (2001). Intra-Amygdala Blockade of the NR2B Subunit of the NMDA Receptor Disrupts the Acquisition But Not the Expression of Fear Conditioning. Journal of Neuroscience, 21(17), 6889-6896.Rossini, P. M., Di Iorio, R., Vecchio, F., Anfossi, M., Babiloni, C., Bozzali, M., Bruni, A. C., Cappa, S. F., Escudero, J., Fraga, F. J., Giannakopoulos, P., Guntekin, B., Logroscino, G., Marra, C., Miraglia, F., Panza, F., Tecchio, F., Pascual-Leone, A., & Dubois, B. (2020). Early diagnosis of Alzheimer’s disease: The role of biomarkers including advanced EEG signal analysis. Report from the IFCN-sponsored panel of experts. Clinical Neurophysiology, 131(6), 1287-1310. https://doi.org/10.1016/j.clinph.2020.03.003Salmon, D. P. (2008). Chapter 5 Neuropsychology of aging and dementia. Handbook of Clinical Neurology, 88, 113-135. Scopus. https://doi.org/10.1016/S0072-9752(07)88005-5Samuraki, M., Matsunari, I., & Yamada, M. (2017). Neuroimaging study of Alzheimer’s disease in volunteer-based cohort. En Neuroimaging Diagnosis for Alzheimer’s Disease and Other Dementias (pp. 243-255). Scopus. https://doi.org/10.1007/978-4-431-55133-1_14Sankari, Z., Adeli, H., & Adeli, A. (2012). Wavelet Coherence Model for Diagnosis of Alzheimer Disease. Clinical EEG and Neuroscience, 43(4), 268-278. https://doi.org/10.1177/1550059412444970Scheltens, P., Leys, D., Barkhof, F., Huglo, D., Weinstein, H. C., Vermersch, P., Kuiper, M., Steinling, M., Wolters, E. C., & Valk, J. (1992). Atrophy of medial temporal lobes on MRI in «probable» Alzheimer’s disease and normal ageing: Diagnostic value and neuropsychological correlates. Journal of Neurology, Neurosurgery, and Psychiatry, 55(10), 967-972. https://doi.org/10.1136/jnnp.55.10.967Schindler, S. E., Bollinger, J. G., Ovod, V., Mawuenyega, K. G., Li, Y., Gordon, B. A., Holtzman, D. M., Morris, J. C., Benzinger, T. L. S., Xiong, C., Fagan, A. M., & Bateman, R. J. (2019). High-precision plasma β-amyloid 42/40 predicts current and future brain amyloidosis. Neurology, 93(17), e1647-e1659. https://doi.org/10.1212/WNL.0000000000008081Scoville, W. B., & Milner, B. (1957). Loss of recent memory after bilateral hippocampal lesions. Journal of neurology, neurosurgery, and psychiatry, 20(1), 11.Sekeres, M. J., Moscovitch, M., & Winocur, G. (2017). Mechanisms of Memory Consolidation and Transformation. En Cognitive Neuroscience of Memory Consolidation (pp. 17-44). Springer.Seppälä, T. T., Nerg, O., Koivisto, A. M., Rummukainen, J., Puli, L., Zetterberg, H., Pyykkö, O. T., Helisalmi, S., Alafuzoff, I., Hiltunen, M., Jääskeläinen, J. E., Rinne, J., Soininen, H., Leinonen, V., & Herukka, S. K. (2012). CSF biomarkers for Alzheimer disease correlate with cortical brain biopsy findings. Neurology, 78(20), 1568-1575. https://doi.org/10.1212/WNL.0b013e3182563bd0Serra, L., Cercignani, M., Lenzi, D., Perri, R., Fadda, L., Caltagirone, C., Macaluso, E., & Bozzali, M. (2010). Grey and white matter changes at different stages of Alzheimer’s disease. Journal of Alzheimer’s Disease: JAD, 19(1), 147-159. https://doi.org/10.3233/JAD-2010-1223Serra, L., Cercignani, M., Petrosini, L., Basile, B., Perri, R., Fadda, L., Spanò, B., Marra, C., Giubilei, F., Carlesimo, G. A., Caltagirone, C., & Bozzali, M. (2011). Neuroanatomical correlates of cognitive reserve in Alzheimer disease. Rejuvenation Research, 14(2), 143-151. https://doi.org/10.1089/rej.2010.1103Shaughnessy, L., Sheard, S., Goldfarb, D., & Atri, A. (2019). Cognitive Assessment of Alzheimer’s Disease and Dementias in Clinical Practice: Pragmatics of Brief Instruments and Neuropsychological Evaluation. The Journal of Clinical Psychiatry, 80(4), MS18002BR2C. https://doi.org/10.4088/JCP.MS18002BR2CSherrington, C. S. (1910). Flexion-reflex of the limb, crossed extension-reflex, and reflex stepping and standing. The Journal of Physiology, 40(1-2), 28-121.Shi, S.-H., Hayashi, Y., Petralia, R. S., Zaman, S. H., Wenthold, R. J., Svoboda, K., & Malinow, R. (1999). Rapid Spine Delivery and Redistribution of AMPA Receptors After Synaptic NMDA Receptor Activation. Science, 284(5421), 1811-1816. https://doi.org/10.1126/science.284.5421.1811Shum, D. H., McFarland, K. A., & Bain, J. D. (1990). Construct validity of eight tests of attention: Comparison of normal and closed head injured samples. The Clinical Neuropsychologist, 4(2), 151-162.Silva, A. J., Kogan, J. H., Frankland, P. W., & Kida, S. (1998). CREB and memory. Annual Review of Neuroscience, 21, 127-148. https://doi.org/10.1146/annurev.neuro.21.1.12Sir Charles Sherrington—Nobel Lecture: Inhibition as a Coordinative Factor. (s. f.). Recuperado 11 de mayo de 2017, de https://www.nobelprize.org/nobel_prizes/medicine/laureates/1932/sherrington lecture.htmlSmall, S. A., Perera, G. M., DeLaPaz, R., Mayeux, R., & Stern, Y. (1999). Differential regional dysfunction of the hippocampal formation among elderly with memory decline and Alzheimer’s disease. Annals of Neurology, 45(4), 466-472. https://doi.org/10.1002/1531-8249(199904)45:4<466::aid-ana8>3.0.co;2-qSmith, G. (2016). Dementia care: A practical approach. CRC Press. https://books.google.com/books?hl=en&lr=&id=6hRjDAAAQBAJ&oi=fnd&pg=PP1&dq=%22Ann+Hayes,+Lorraine+Shaw,+and+Grahame%22+%22Psychological+interventions+in%22+%22Parker+and+Robert+G.%22+%22Smith+and+Denise%22+%22Rylance+and+Donal%22+%22End+of+life%22+%22Knott+and+Denise%22+&ots=2g0s9zCAMk&sig=pE__72LTF6oiZoKm4x5-3CR3vosSoltani Zangbar, H., Soltani Zangbar, H., Ghadiri, T., Seyedi Vafaee, M., Ebrahimi Kalan, A., Fallahi, S., Ghorbani, M., & Shahabi, P. (2020). Theta Oscillations through Hippocampal/Prefrontal Pathway: Importance in Cognitive Performances. Brain Connectivity, 10(4), 157-169. Scopus. https://doi.org/10.1089/brain.2019.0733Sperling, R. A., Bates, J. F., Chua, E. F., Cocchiarella, A. J., Rentz, D. M., Rosen, B. R., Schacter, D. L., & Albert, M. S. (2003). FMRI studies of associative encoding in young and elderly controls and mild Alzheimer’s disease. Journal of Neurology, Neurosurgery, and Psychiatry, 74(1), 44-50. https://doi.org/10.1136/jnnp.74.1.44Squire, L. R. (1992). Memory and the hippocampus: A synthesis from findings with rats, monkeys, and humans. Psychological Review, 99(2), 195-231.Stam, C. J., van Cappellen van Walsum, A. M., Pijnenburg, Y. A. L., Berendse, H. W., de Munck, J. C., Scheltens, P., & van Dijk, B. W. (2002). Generalized Synchronization of MEG Recordings in Alzheimer’s Disease: Evidence for Involvement of the Gamma Band. Journal of Clinical Neurophysiology, 19(6), 562-574.Stam, C. J., van der Made, Y., Pijnenburg, Y. a. L., & Scheltens, P. (2003). EEG synchronization in mild cognitive impairment and Alzheimer’s disease. Acta Neurologica Scandinavica, 108(2), 90-96. https://doi.org/10.1034/j.1600-0404.2003.02067.xStanton, P. K., & Sarvey, J. M. (1984). Blockade of long-term potentiation in rat hippocampal CA1 region by inhibitors of protein synthesis. Journal of Neuroscience, 4(12), 3080-3088.Stefanowska, M. (1897). Les appendices terminaux des dendrites cérébraux et leurs différents états physiologiques. Hayez, Imprimeur de l’Académie royale des sciences, des lettres et des beaux-arts de Belgique.Stomrud, E., Hansson, O., Minthon, L., Blennow, K., Rosén, I., & Londos, E. (2010). Slowing of EEG correlates with CSF biomarkers and reduced cognitive speed in elderly with normal cognition over 4 years. Neurobiology of Aging, 31(2), 215-223. https://doi.org/10.1016/j.neurobiolaging.2008.03.025Strauss, E., Sherman, E. M., & Spreen, O. (2006). A compendium of neuropsychological tests: Administration, norms, and commentary. American chemical society.Suárez-Calvet, M., Karikari, T. K., Ashton, N. J., Lantero Rodríguez, J., Milà-Alomà, M., Gispert, J. D., Salvadó, G., Minguillon, C., Fauria, K., Shekari, M., Grau-Rivera, O., Arenaza-Urquijo, E. M., Sala-Vila, A., Sánchez-Benavides, G., González-de Echávarri, J. M., Kollmorgen, G., Stoops, E., Vanmechelen, E., Zetterberg, H., … ALFA Study. (2020). Novel tau biomarkers phosphorylated at T181, T217 or T231 rise in the initial stages of the preclinical Alzheimer’s continuum when only subtle changes in Aβ pathology are detected. EMBO Molecular Medicine, 12(12), e12921. https://doi.org/10.15252/emmm.202012921Tabaton, M., Nunzi, M. G., Xue, R., Usiak, M., Autilio-Gambetti, L., & Gambetti, P. (1994). Soluble amyloid beta-protein is a marker of Alzheimer amyloid in brain but not in cerebrospinal fluid. Biochemical and Biophysical Research Communications, 200(3), 1598-1603. https://doi.org/10.1006/bbrc.1994.1634Tang, Y. P., Wang, H., Feng, R., Kyin, M., & Tsien, J. Z. (2001). Differential effects of enrichment on learning and memory function in NR2B transgenic mice. Neuropharmacology, 41(6), 779-790.Tangalos, E. G., & Petersen, R. C. (2018). Mild Cognitive Impairment in Geriatrics. Clinics in Geriatric Medicine, 34(4), 563-589. Scopus. https://doi.org/10.1016/j.cger.2018.06.005Tapiola, T., Alafuzoff, I., Herukka, S.-K., Parkkinen, L., Hartikainen, P., Soininen, H., & Pirttilä, T. (2009). Cerebrospinal fluid {beta}-amyloid 42 and tau proteins as biomarkers of Alzheimer-type pathologic changes in the brain. Archives of Neurology, 66(3), 382-389. https://doi.org/10.1001/archneurol.2008.596Tatebe, H., Kasai, T., Ohmichi, T., Kishi, Y., Kakeya, T., Waragai, M., Kondo, M., Allsop, D., & Tokuda, T. (2017). Quantification of plasma phosphorylated tau to use as a biomarker for brain Alzheimer pathology: Pilot case-control studies including patients with Alzheimer’s disease and down syndrome. Molecular Neurodegeneration, 12(1), 63. https://doi.org/10.1186/s13024-017-0206-8The Alzheimer’s Association QC program for CSF and blood biomarkers | University of Gothenburg. (s. f.). Recuperado 11 de marzo de 2022, de https://www.gu.se/en/neuroscience-physiology/the-alzheimers-association-qc program-for-csf-and-blood-biomarkerThiyagesh, S. N., Farrow, T. F. D., Parks, R. W., Accosta-Mesa, H., Young, C., Wilkinson, I. D., Hunter, M. D., & Woodruff, P. W. R. (2009). The neural basis of visuospatial perception in Alzheimer’s disease and healthy elderly comparison subjects: An fMRI study. Psychiatry Research, 172(2), 109-116. https://doi.org/10.1016/j.pscychresns.2008.11.002Tononi, G., Edelman, G. M., & Sporns, O. (1998). Complexity and coherency: Integrating information in the brain. Trends in Cognitive Sciences, 2(12), 474-484. https://doi.org/10.1016/S1364-6613(98)01259-5Tononi, G., Sporns, O., & Edelman, G. M. (1994). A measure for brain complexity: Relating functional segregation and integration in the nervous system. Proceedings of the National Academy of Sciences of the United States of America, 91(11), 5033-5037. https://doi.org/10.1073/pnas.91.11.5033Torisson, G., van Westen, D., Stavenow, L., Minthon, L., & Londos, E. (2015). Medial temporal lobe atrophy is underreported and may have important clinical correlates in medical inpatients. BMC Geriatrics, 15(1), 65. https://doi.org/10.1186/s12877-015-0066-4Tse, D., Takeuchi, T., Kakeyama, M., Kajii, Y., Okuno, H., Tohyama, C., Bito, H., & Morris, R. G. (2011). Schema-dependent gene activation and memory encoding in neocortex. Science, 333(6044), 891-895.Tulving, E. (1972). Episodic and semantic memory.Tzourio-Mazoyer, N., Landeau, B., Papathanassiou, D., Crivello, F., Etard, O., Delcroix, N., Mazoyer, B., & Joliot, M. (2002). Automated Anatomical Labeling of Activations in SPM Using a Macroscopic Anatomical Parcellation of the MNI MRI Single-Subject Brain. NeuroImage, 15(1), 273-289.https://doi.org/10.1006/nimg.2001.0978van den Heuvel, M. P., & Hulshoff Pol, H. E. (2010). Exploring the brain network: A review on resting-state fMRI functional connectivity. European Neuropsychopharmacology, 20(8), 519-534. Scopus. https://doi.org/10.1016/j.euroneuro.2010.03.008van der Hiele, K., Vein, A. A., Reijntjes, R. H. a. M., Westendorp, R. G. J., Bollen, E. L. E. M., van Buchem, M. A., van Dijk, J. G., & Middelkoop, H. a. M. (2007). EEG correlates in the spectrum of cognitive decline. Clinical Neurophysiology: Official Journal of the International Federation of Clinical Neurophysiology, 118(9), 1931-1939. https://doi.org/10.1016/j.clinph.2007.05.070van Kesteren, M. T., Fernández, G., Norris, D. G., & Hermans, E. J. (2010). Persistent schema-dependent hippocampal-neocortical connectivity during memory encoding and postencoding rest in humans. Proceedings of the National Academy of Sciences, 107(16), 7550-7555.van Kesteren, M. T., Rijpkema, M., Ruiter, D. J., & Fernández, G. (2010). Retrieval of associative information congruent with prior knowledge is related to increased medial prefrontal activity and connectivity. Journal of Neuroscience, 30(47), 15888-15894.VanderWeele, T. J. (2019). Principles of confounder selection. European Journal of Epidemiology, 34(3), 211-219. https://doi.org/10.1007/s10654-019-00494-6Vanhoenacker, A.-S., Sneyers, B., De Keyzer, F., Heye, S., & Demaerel, P. (2017). Evaluation and clinical correlation of practical cut-offs for visual rating scales of atrophy: Normal aging versus mild cognitive impairment and Alzheimer’s disease. Acta Neurologica Belgica, 117(3), 661-669. Scopus. https://doi.org/10.1007/s13760-017-0777-8Varela, F., Lachaux, J.-P., Rodriguez, E., & Martinerie, J. (2001). The brainweb: Phase synchronization and large-scale integration. Nature Reviews Neuroscience, 2(4), Art. 4. https://doi.org/10.1038/35067550Vecchio, F., Miraglia, F., Iberite, F., Lacidogna, G., Guglielmi, V., Marra, C., Pasqualetti, P., Tiziano, F. D., & Rossini, P. M. (2018). Sustainable method for Alzheimer dementia prediction in mild cognitive impairment: Electroencephalographic connectivity and graph theory combined with apolipoprotein E: MCI Conversion. Annals of Neurology, 84(2), 302-314. https://doi.org/10.1002/ana.25289Vecchio, F., Miraglia, F., Judica, E., Cotelli, M., Alù, F., & Rossini, P. M. (2020). Human brain networks: A graph theoretical analysis of cortical connectivity normative database from EEG data in healthy elderly subjects. GeroScience, 42(2), 575-584. https://doi.org/10.1007/s11357-020-00176-2Vermunt, L., Sikkes, S. A. M., van den Hout, A., Handels, R., Bos, I., van der Flier, W. M., Kern, S., Ousset, P.-J., Maruff, P., Skoog, I., Verhey, F. R. J., Freund-Levi, Y., Tsolaki, M., Wallin, Å. K., Olde Rikkert, M., Soininen, H., Spiru, L., Zetterberg, H., Blennow, K., … ICTUS/DSA study groups. (2019). Duration of preclinical, prodromal, and dementia stages of Alzheimer’s disease in relation to age, sex, and APOE genotype. Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association, 15(7), 888-898. https://doi.org/10.1016/j.jalz.2019.04.001Wahlund, L.-O., Westman, E., van Westen, D., Wallin, A., Shams, S., Cavallin, L., Larsson, E.-M., & From, the I. C. I. N. (ICINET). (2017). Imaging biomarkers of dementia: Recommended visual rating scales with teaching cases. Insights into Imaging, 8(1), 79-90. Scopus. https://doi.org/10.1007/s13244-016-0521-6Wang, R., Wang, J., Yu, H., Wei, X., Yang, C., & Deng, B. (2015). Power spectral density and coherence analysis of Alzheimer’s EEG. Cognitive Neurodynamics, 9(3), 291-304. https://doi.org/10.1007/s11571-014-9325-xWanleenuwat, P., Iwanowski, P., & Kozubski, W. (2019). Alzheimer’s dementia: Pathogenesis and impact of cardiovascular risk factors on cognitive decline. Postgraduate Medicine, 131(7), 415-422. Scopus. https://doi.org/10.1080/00325481.2019.1657776Welle (www.dw.com), D. (s. f.). UNESCO: Dos de cada diez personas en América Latina no tienen los niveles mínimos de compresión de lectura | DW | 06.09.2019. DW.COM. Recuperado 12 de marzo de 2022, de https://www.dw.com/es/unesco-dos-de-cada diez-personas-en-am%C3%A9rica-latina-no-tienen-los-niveles m%C3%ADnimos de-compresi%C3%B3n-de-lectura/a-50333467Weltgesundheitsorganisation, & Alzheimer’s Disease International (Eds.). (2012). Dementia: A public health priority.Willis, T. (1962). 1664. Cerebri anatome, cui accessit nervorum descriptio et usus. London: Martin & Allestry by University College London on, 5(09), 06.Wysokiński, A., Zboralski, K., Orzechowska, A., Gałecki, P., Florkowski, A., & Talarowska, M. (2010). Normalization of the Verbal Fluency Test on the basis of results for healthy subjects, patients with schizophrenia, patients with organic lesions of the chronic nervous system and patients with type 1 and 2 diabetes. Archives of Medical Science, 6(3), 438-446.Yetkin, F. Z., Rosenberg, R. N., Weiner, M. F., Purdy, P. D., & Cullum, C. M. (2006). FMRI of working memory in patients with mild cognitive impairment and probable Alzheimer’s disease. European Radiology, 16(1), 193-206. https://doi.org/10.1007/s00330-005-2794-xYi, H.-A., Won, K. S., Chang, H. W., & Kim, H. W. (2018). Association between white matter lesions and cerebral Aβ burden. PLoS ONE, 13(9). Scopus. https://doi.org/10.1371/journal.pone.0204313Yin, Y., Edelman, G. M., & Vanderklish, P. W. (2002). The brain-derived neurotrophic factor enhances synthesis of Arc in synaptoneurosomes. Proceedings of the National Academy of Sciences, 99(4), 2368-2373. https://doi.org/10.1073/pnas.042693699Yuste, R. (2010). Dendritic Spines. MIT Press.Zegarra-Valdivia, J., Solís, L. D., & Chino-Vilca, B. (2019). Effectiveness of the photo-test front of the MMSE, for the screening of cognitive deterioration in Peruvian population. Revista Ecuatoriana de Neurologia, 28(1), 39-46. Scopus.Zetterberg, H. (2016). Neurofilament Light: A Dynamic Cross-Disease Fluid Biomarker for Neurodegeneration. Neuron, 91(1), 1-3. https://doi.org/10.1016/j.neuron.2016.06.030Zetterberg, H., Skillbäck, T., Mattsson, N., Trojanowski, J. Q., Portelius, E., Shaw, L. M., Weiner, M. W., Blennow, K., & Alzheimer’s Disease Neuroimaging Initiative. (2016). Association of Cerebrospinal Fluid Neurofilament Light Concentration With Alzheimer Disease Progression. JAMA Neurology, 73(1), 60-67. https://doi.org/10.1001/jamaneurol.2015.3037Zhang, H., & Jacobs, J. (2015). Traveling theta waves in the human hippocampus. Journal of Neuroscience, 35(36), 12477-12487.Demencia tipo alzheimer (DA)Deterioro cognitivo leve (DCL)Electroencefalograma (EEG)Espectro de potenciaConectividad funcionalFototestMoCAAlzheimer's dementia (AD)Mild cognitive impairment (MCI)Electroencephalogram (EEG)Power spectraFunctional connectivityORIGINALPerfil de conectividad cerebral y el rendimiento cognitivo en consulta.pdfPerfil de conectividad cerebral y el rendimiento cognitivo en consulta.pdfTesisapplication/pdf162774https://repositorio.cuc.edu.co/bitstream/11323/10766/1/Perfil%20de%20conectividad%20cerebral%20y%20el%20rendimiento%20cognitivo%20en%20consulta.pdff581aa4152a2081208cc2eb41bb9707dMD51open accessLICENSElicense.txtlicense.txttext/plain; charset=utf-814828https://repositorio.cuc.edu.co/bitstream/11323/10766/2/license.txt2f9959eaf5b71fae44bbf9ec84150c7aMD52open accessTEXTPerfil de conectividad cerebral y el rendimiento cognitivo en consulta.pdf.txtPerfil de conectividad cerebral y el rendimiento cognitivo en consulta.pdf.txtExtracted texttext/plain3618https://repositorio.cuc.edu.co/bitstream/11323/10766/3/Perfil%20de%20conectividad%20cerebral%20y%20el%20rendimiento%20cognitivo%20en%20consulta.pdf.txtc4e7ee6ed4877b5f9fdb5b641c9554c0MD53open accessTHUMBNAILPerfil de conectividad cerebral y el rendimiento cognitivo en consulta.pdf.jpgPerfil de conectividad cerebral y el rendimiento cognitivo en consulta.pdf.jpgGenerated Thumbnailimage/jpeg6602https://repositorio.cuc.edu.co/bitstream/11323/10766/4/Perfil%20de%20conectividad%20cerebral%20y%20el%20rendimiento%20cognitivo%20en%20consulta.pdf.jpg68106b15834b5c5cd4a2253177585e76MD54open access11323/10766oai:repositorio.cuc.edu.co:11323/107662024-02-22 03:01:14.048An error occurred on the license name.|||https://creativecommons.org/licenses/by-nc-sa/4.0/open accessRepositorio Universidad de La Costabdigital@metabiblioteca.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