1
|
Giraldo-Berrio D, Jimenez-Del-Rio M, Velez-Pardo C. Minocycline mitigates Aβ and TAU pathology, neuronal dysfunction, and death in the PSEN1 E280A cholinergic-like neurons model of familial Alzheimer's disease. Neuropharmacology 2024; 261:110152. [PMID: 39245141 DOI: 10.1016/j.neuropharm.2024.110152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Familial Alzheimer's disease (FAD) presenilin 1 E280A (PSEN1 E280A) is a severe neurological condition due to the loss of cholinergic neurons (ChNs), accumulation of amyloid beta (Aβ), and abnormal phosphorylation of the TAU protein. Up to date, there are no effective therapies available. The need for innovative treatments for this illness is critical. We found that minocycline (MC, 5 μM) was innocuous toward wild-type (WT) PSEN1 ChLNs but significantly (i) reduces the accumulation of intracellular Aβ by -69%, (ii) blocks both abnormal phosphorylation of the protein TAU at residue Ser202/Thr205 by -33% and (iii) phosphorylation of the proapoptotic transcription factor c-JUN at residue Ser63/Ser73 by -25%, (iv) diminishes oxidized DJ-1 at Cys106-SO3 by -29%, (v) downregulates the expression of transcription factor TP53, (vi) BH-3-only protein PUMA, and (vii) cleaved caspase 3 (CC3) by -33, -86, and -78%, respectively, compared with untreated PSEN1 E280A ChLNs. Additionally, MC increases the response to ACh-induced Ca2+ influx by +92% in mutant ChLNs. Oxygen radical absorbance capacity (ORAC) and ferric ion-reducing antioxidant power (FRAP) analysis showed that MC might operate more efficiently as a hydrogen atom transfer agent than a single electron transfer agent. In silico molecular docking analysis predicts that MC binds with high affinity to Aβ (Vina Score -6.6 kcal/mol), TAU (VS -6.5 kcal/mol), and caspase 3 (VS -7.1 kcal/mol). Taken together, our findings suggest that MC demonstrates antioxidant, anti-amyloid, and anti-apoptosis activity and promotes physiological ACh-induced Ca2+ influx in PSEN1 E280A ChLNs. The MC has therapeutic potential for treating early-onset FAD.
Collapse
Affiliation(s)
- Daniela Giraldo-Berrio
- Neuroscience Research Group, Institute of Medical Investigations, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Torre 1, Laboratory 412, Medellín, Colombia
| | - Marlene Jimenez-Del-Rio
- Neuroscience Research Group, Institute of Medical Investigations, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Torre 1, Laboratory 412, Medellín, Colombia.
| | - Carlos Velez-Pardo
- Neuroscience Research Group, Institute of Medical Investigations, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Torre 1, Laboratory 412, Medellín, Colombia.
| |
Collapse
|
2
|
Bustamante C, Martinez JF, Navarro A, Lopera M, Villegas G, Duque S, Acosta-Baena N, Ríos-Romenets S, Lopera F. Strategies to promote contraception use by female volunteers in Alzheimer's Prevention Initiative Autosomal-Dominant Alzheimer's Disease (API ADAD) Colombia trial. Clin Trials 2024:17407745241264217. [PMID: 39143683 DOI: 10.1177/17407745241264217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
BACKGROUND/AIMS Including women of childbearing age in a clinical trial makes it necessary to consider two factors from a bioethical perspective: first, the lack of knowledge about the potential teratogenic effects of an investigational product, and also, the principle of justice not to exclude any population from the benefits of research. The most common way to address this issue is by requiring volunteers to use contraceptives before, during, and a few weeks after the clinical trial. This work presents all the strategies used to promote contraception use and prevent pregnancy during the Alzheimer's Prevention Initiative Autosomal-Dominant Alzheimer's Disease (API ADAD) Colombia clinical trial. Two characteristics of this trial make it of special interest for closely monitoring contraception use. One is that the trial lasted more than 7 years, and the other is that participants could be carriers of the E280A PSEN1 mutation, leading to a mild cognitive impairment as early as their late 30s. METHODS An individual medical evaluation to select the contraception method that best fits the volunteer was carried out during the screening visit, remitting to the gynecologist when necessary. All non-surgical contraception methods were supplied by the sponsor. Staff were trained on contraception counseling, correctly dispensing contraceptive drugs to volunteers, and identifying, reporting, and following up on pregnancies. Two comprehensive educational campaigns on contraception use were performed, and the intervention included all volunteers. In addition, volunteers were asked on an annual survey to evaluate the dispensing procedure. Finally, the effectiveness of these strategies was retrospectively evaluated, comparing by extrapolation the number of pregnancies presented throughout the trial with the General Fertility Rate in Colombia. RESULTS A total of 159 female volunteers were recruited. All strategies were implemented as planned, even during the COVID-19 contingency. Ten pregnancies occurred during the evaluation period (2015-2021). Two were planned; the rest were associated with a potential therapeutic failure or incorrect use of contraceptive methods for a contraceptive failure of 0.49% per year. Sixty percent of pregnancies led to an abortion, either miscarriage or therapeutic abortion. However, there was not enough data to associate the pregnancy outcome with the administration of the investigational product. Finally, we observed a lower fertility rate in women participating in the trial compared to the Colombian population. CONCLUSION The lower rates of contraceptive failure and the decrease in the incidence of pregnancies in women participating in the trial compared to the Colombian population across the 7 years of evaluation suggest that the strategies used in API ADAD Colombia were adequate and effective in addressing contraception use.
Collapse
Affiliation(s)
| | - Juan F Martinez
- Neurosciences Group of Antioquia, University of Antioquia, Medellin, Colombia
| | - Alexander Navarro
- Neurosciences Group of Antioquia, University of Antioquia, Medellin, Colombia
| | - Margarita Lopera
- Neurosciences Group of Antioquia, University of Antioquia, Medellin, Colombia
| | - Gustavo Villegas
- Neurosciences Group of Antioquia, University of Antioquia, Medellin, Colombia
| | - Sindy Duque
- Neurosciences Group of Antioquia, University of Antioquia, Medellin, Colombia
| | | | | | - Francisco Lopera
- Neurosciences Group of Antioquia, University of Antioquia, Medellin, Colombia
| |
Collapse
|
3
|
Huq A, Thompson B, Winship I. Clinical application of whole genome sequencing in young onset dementia: challenges and opportunities. Expert Rev Mol Diagn 2024; 24:659-675. [PMID: 39135326 DOI: 10.1080/14737159.2024.2388765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/01/2024] [Indexed: 08/30/2024]
Abstract
INTRODUCTION Young onset dementia (YOD) by its nature is difficult to diagnose. Despite involvement of multidisciplinary neurogenetics services, patients with YOD and their families face significant diagnostic delays. Genetic testing for people with YOD currently involves a staggered, iterative approach. There is currently no optimal single genetic investigation that simultaneously identifies the different genetic variants resulting in YOD. AREAS COVERED This review discusses the advances in clinical genomic testing for people with YOD. Whole genome sequencing (WGS) can be employed as a 'one stop shop' genomic test for YOD. In addition to single nucleotide variants, WGS can reliably detect structural variants, short tandem repeat expansions, mitochondrial genetic variants as well as capture single nucleotide polymorphisms for the calculation of polygenic risk scores. EXPERT OPINION WGS, when used as the initial genetic test, can enhance the likelihood of a precision diagnosis and curtail the time taken to reach this. Finding a clinical diagnosis using WGS can reduce invasive and expensive investigations and could be cost effective. These advances need to be balanced against the limitations of the technology and the genetic counseling needs for these vulnerable patients and their families.
Collapse
Affiliation(s)
- Aamira Huq
- Department of Genomic Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Bryony Thompson
- Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Pathology, University of Melbourne, Parkville, Victoria, Australia
| | - Ingrid Winship
- Department of Genomic Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
4
|
Sosa AL, Brucki SMD, Crivelli L, Lopera FJ, Acosta DM, Acosta‐Uribe J, Aguilar D, Aguilar‐Navarro SG, Allegri RF, Bertolucci PHF, Calandri IL, Carrillo MC, Mendez PAC, Cornejo‐Olivas M, Custodio N, Damian A, de Souza LC, Duran‐Aniotz C, García AM, García‐Peña C, Gonzales MM, Grinberg LT, Ibanez AM, Illanes‐Manrique MZ, Jack CR, Leon‐Salas JM, Llibre‐Guerra JJ, Luna‐Muñoz J, Matallana D, Miller BL, Naci L, Parra MA, Pericak‐Vance M, Piña‐Escudero SD, França Resende EDP, Ringman JM, Sevlever G, Slachevsky A, Suemoto CK, Valcour V, Villegas‐Lanau A, Yassuda MS, Mahinrad S, Sexton C. Advancements in dementia research, diagnostics, and care in Latin America: Highlights from the 2023 Alzheimer's Association International conference satellite symposium in Mexico City. Alzheimers Dement 2024; 20:5009-5026. [PMID: 38801124 PMCID: PMC11247679 DOI: 10.1002/alz.13850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/03/2024] [Indexed: 05/29/2024]
Abstract
INTRODUCTION While Latin America (LatAm) is facing an increasing burden of dementia due to the rapid aging of the population, it remains underrepresented in dementia research, diagnostics, and care. METHODS In 2023, the Alzheimer's Association hosted its eighth satellite symposium in Mexico, highlighting emerging dementia research, priorities, and challenges within LatAm. RESULTS Significant initiatives in the region, including intracountry support, showcased their efforts in fostering national and international collaborations; genetic studies unveiled the unique genetic admixture in LatAm; researchers conducting emerging clinical trials discussed ongoing culturally specific interventions; and the urgent need to harmonize practices and studies, improve diagnosis and care, and use affordable biomarkers in the region was highlighted. DISCUSSION The myriad of topics discussed at the 2023 AAIC satellite symposium highlighted the growing research efforts in LatAm, providing valuable insights into dementia biology, genetics, epidemiology, treatment, and care.
Collapse
|
5
|
Kalaria R, Maestre G, Mahinrad S, Acosta DM, Akinyemi RO, Alladi S, Allegri RF, Arshad F, Babalola DO, Baiyewu O, Bak TH, Bellaj T, Brodie‐Mends DK, Carrillo MC, Celestin K, Damasceno A, de Silva RK, de Silva R, Djibuti M, Dreyer AJ, Ellajosyula R, Farombi TH, Friedland RP, Garza N, Gbessemehlan A, Georgiou EE, Govia I, Grinberg LT, Guerchet M, Gugssa SA, Gumikiriza‐Onoria JL, Hogervorst E, Hornberger M, Ibanez A, Ihara M, Issac TG, Jönsson L, Karanja WM, Lee JH, Leroi I, Livingston G, Manes FF, Mbakile‐Mahlanza L, Miller BL, Musyimi CW, Mutiso VN, Nakasujja N, Ndetei DM, Nightingale S, Novotni G, Nyamayaro P, Nyame S, Ogeng'o JA, Ogunniyi A, de Oliveira MO, Okubadejo NU, Orrell M, Paddick S, Pericak‐Vance MA, Pirtosek Z, Potocnik FCV, Raman R, Rizig M, Rosselli M, Salokhiddinov M, Satizabal CL, Sepulveda‐Falla D, Seshadri S, Sexton CE, Skoog I, George‐Hyslop PHS, Suemoto CK, Thapa P, Udeh‐Momoh CT, Valcour V, Vance JM, Varghese M, Vera JH, Walker RW, Zetterberg H, Zewde YZ, Ismail O. The 2022 symposium on dementia and brain aging in low- and middle-income countries: Highlights on research, diagnosis, care, and impact. Alzheimers Dement 2024; 20:4290-4314. [PMID: 38696263 PMCID: PMC11180946 DOI: 10.1002/alz.13836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 05/04/2024]
Abstract
Two of every three persons living with dementia reside in low- and middle-income countries (LMICs). The projected increase in global dementia rates is expected to affect LMICs disproportionately. However, the majority of global dementia care costs occur in high-income countries (HICs), with dementia research predominantly focusing on HICs. This imbalance necessitates LMIC-focused research to ensure that characterization of dementia accurately reflects the involvement and specificities of diverse populations. Development of effective preventive, diagnostic, and therapeutic approaches for dementia in LMICs requires targeted, personalized, and harmonized efforts. Our article represents timely discussions at the 2022 Symposium on Dementia and Brain Aging in LMICs that identified the foremost opportunities to advance dementia research, differential diagnosis, use of neuropsychometric tools, awareness, and treatment options. We highlight key topics discussed at the meeting and provide future recommendations to foster a more equitable landscape for dementia prevention, diagnosis, care, policy, and management in LMICs. HIGHLIGHTS: Two-thirds of persons with dementia live in LMICs, yet research and costs are skewed toward HICs. LMICs expect dementia prevalence to more than double, accompanied by socioeconomic disparities. The 2022 Symposium on Dementia in LMICs addressed advances in research, diagnosis, prevention, and policy. The Nairobi Declaration urges global action to enhance dementia outcomes in LMICs.
Collapse
Grants
- P30AG066506 National Institute of Aging (NIA)
- P01 HD035897 NICHD NIH HHS
- R13 AG066391 NIA NIH HHS
- International Society for Neurochemistry
- National Council for Scientific and Technological Development
- R01 AG075775 NIA NIH HHS
- Bluefield Project, the Olav Thon Foundation, the Erling-Persson Family Foundation, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden
- U19 AG074865 NIA NIH HHS
- UH3 NS100605 NINDS NIH HHS
- R01AG072547 Multi partner Consortium for Dementia Research in Latino America-Dominican Republic (LATAM-FINGERS)
- ASP/06/RE/2012/18 University of Sri Jayewardenepura, Sri Lanka
- D43 TW011532 FIC NIH HHS
- UF1 NS125513 NINDS NIH HHS
- 2019-02397 Swedish Research Council
- FLR/R1/191813 UK Royal Society/African Academy of Sciences
- R01 AG054076 NIA NIH HHS
- GOK: Government of Karnataka
- R56 AG074467 NIA NIH HHS
- R21 AG069252 NIA NIH HHS
- RF1 AG059421 NIA NIH HHS
- R56 AG061837 NIA NIH HHS
- Global Brain Health Institute (GBHI)
- 1R01AG068472-01 National Institute of Aging (NIA)
- FCG/R1/201034 UK Royal Society/African Academy of Sciences
- Appel à Projet des Equipes Émergentes et Labellisées scheme (APREL)
- Alzheimer's Drug Discovery Foundation (ADDF)
- R01 AG062588 NIA NIH HHS
- 1R01AG070883 University of Wisconsin, Madison
- U01 HG010273 NHGRI NIH HHS
- R25 TW011214 FIC NIH HHS
- ASP/06/RE/2013/28 University of Sri Jayewardenepura, Sri Lanka
- R01 AG052496 NIA NIH HHS
- R01 AG080468 NIA NIH HHS
- RBM: Rotary Bangalore Midtown
- U19 AG068054 NIA NIH HHS
- ADSF-21-831376-C Alzheimer Drug Discovery Foundation
- ADSF-21-831377-C Alzheimer Drug Discovery Foundation
- Canadian Institute of Health Research
- U19 AG078558 NIA NIH HHS
- 1P30AG066546-01A1 National Institutes of Health (NIH)
- RF1 AG059018 NIA NIH HHS
- National Research Foundation (NRF)
- P30 AG062422 NIA NIH HHS
- LSIPL: M/s Lowes Services India Private Limited
- UKDRI-1003 UK Dementia Research Institute at UCL
- U19AG074865 Multi partner Consortium for Dementia Research in Latino America-Dominican Republic (LATAM-FINGERS)
- P01 AG019724 NIA NIH HHS
- National Institute for Health and Care Research, United Kingdom
- R01 AG066524 NIA NIH HHS
- RF1 AG063507 NIA NIH HHS
- WCUP/Ph.D./19B 2013 University of Sri Jayewardenepura (USJ), Sri Lanka
- WCUP/Ph.D./19/2013 University of Sri Jayewardenepura (USJ), Sri Lanka
- GBHI ALZ UK-21-724359 Pilot Award for Global Brain Health Leaders
- R01AG080468-01 National Institute of Aging (NIA)
- U01 AG058589 NIA NIH HHS
- R01 AG057234 NIA NIH HHS
- SP/CIN/2016/02) Ministry of Primary Industries, Sri Lanka
- R01 AG072547 NIA NIH HHS
- U01 AG051412 NIA NIH HHS
- P30 AG059305 NIA NIH HHS
- Alzheimer's Association, USA
- R35 AG072362 NIA NIH HHS
- R01 NS050915 NINDS NIH HHS
- P30 AG066546 NIA NIH HHS
- 2022-01018 Swedish Research Council
- U19 AG063893 NIA NIH HHS
- ALFGBG-71320 Swedish State Support for Clinical Research
- U01 AG052409 NIA NIH HHS
- 1R13AG066391-01 National Institutes of Health (NIH)
- R01 AG21051 NIH and the Fogarty International Center [FIC]
- DP1AG069870 National Institutes of Health (NIH)
- Marie Skłodowska-Curie
- U19 AG078109 NIA NIH HHS
- Chinese Neuroscience Society, China
- RF1 AG061872 NIA NIH HHS
- DP1 AG069870 NIA NIH HHS
- P30 AG066506 NIA NIH HHS
- Wellcome Trust
- U01HG010273 Multi partner Consortium for Dementia Research in Latino America-Dominican Republic (LATAM-FINGERS)
- JPND2021-00694 European Union Joint Programme - Neurodegenerative Disease Research
- ASP/06/RE/2010/07 University of Sri Jayewardenepura, Sri Lanka
- Rainwater Charitable Foundation - The Bluefield project to cure FTD, and Global Brain Health Institute
- 101053962 European Union's Horizon Europe
- R01 AG058464 NIA NIH HHS
- R01 AG068472 NIA NIH HHS
- Michael J. Fox Foundation for Parkinson's Research, USA
- UL1 TR001873 NCATS NIH HHS
- SG-21-814756 National Institutes of Health (NIH)
- 201809-2016862 Alzheimer Drug Discovery Foundation
- UK National Health Service, Newcastle University,
- R01 AG058918 NIA NIH HHS
- National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
- Wellcome Trust, UK
- ADSF-21-831381-C Alzheimer Drug Discovery Foundation
- Health Professionals Education Partnership Initiative Ethiopia
- ANR-09-MNPS-009-01 French National Research Agency
- R01 AG062562 NIA NIH HHS
- AXA Research Fund
- ICMR: Indian Council for Medical Research
- R01 AG070883 NIA NIH HHS
- International Society for Neurochemistry
- French National Research Agency
- AXA Research Fund
- National Center for Advancing Translational Sciences
- National Council for Scientific and Technological Development
- Swedish Research Council
Collapse
Affiliation(s)
- Raj Kalaria
- Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Gladys Maestre
- Departments of Neuroscience and Human GeneticsUniversity of Texas Rio Grande ValleyOne W. University BlvdBrownsvilleTexasUSA
| | - Simin Mahinrad
- Division of Medical and Scientific RelationsAlzheimer's AssociationChicagoIllinoisUSA
| | - Daisy M. Acosta
- Universidad Nacional Pedro Henriquez Urena (UNPHU)Santo DomingoDominican Republic
| | - Rufus Olusola Akinyemi
- Neuroscience and Ageing Research UnitInstitute for Advanced Medical Research and TrainingCollege of MedicineUniversity of IbadanIbadanOyoNigeria
| | - Suvarna Alladi
- Department of NeurologyNational Institute of Mental Health and NeurosciencesBengaluruKarnatakaIndia
| | - Ricardo F. Allegri
- Fleni Neurological InstituteBuenos AiresArgentina
- Department of NeurosciencesUniversidad de la Costa (CUC)BarranquillaColombia
| | - Faheem Arshad
- Department of NeurologyNational Institute of Mental Health and NeurosciencesBengaluruKarnatakaIndia
| | | | | | | | | | | | - Maria C. Carrillo
- Division of Medical and Scientific RelationsAlzheimer's AssociationChicagoIllinoisUSA
| | - Kaputu‐Kalala‐Malu Celestin
- Department of NeurologyCentre Neuropsychopathologique (CNPP)Kinshasa University Teaching HospitalUniversity of KinshasaKinshasaRepublic Democratic of the Congo
| | | | - Ranil Karunamuni de Silva
- Interdisciplinary Centre for Innovation in Biotechnology and NeuroscienceFaculty of Medical SciencesUniversity of Sri JayewardenepuraNugegodaSri Lanka
- Institute for Combinatorial Advanced Research and Education (KDU‐CARE)General Sir John Kotelawala Defence UniversityRatmalanaSri Lanka
| | - Rohan de Silva
- Reta Lila Weston Institute and Department of ClinicalMovement NeuroscienceUCL Queen Square Institute of NeurologyLondonUK
| | - Mamuka Djibuti
- Partnership for Research and Action for Health (PRAH)TbilisiGeorgia
| | | | - Ratnavalli Ellajosyula
- Cognitive Neurology ClinicManipal Hospitaland Annasawmy Mudaliar HospitalBengaluruKarnatakaIndia
- Manipal Academy of Higher Education (MAHE)ManipalKarnatakaIndia
| | | | | | - Noe Garza
- Department of Neuroscience and Human GeneticsUniversity of Texas Rio Grande ValleyHarlingenTexasUSA
| | - Antoine Gbessemehlan
- Inserm U1094, IRD U270University of LimogesCHU Limoges, EpiMaCT ‐ Epidemiology of Chronic Diseases in Tropical ZoneInstitute of Epidemiology and Tropical NeurologyOmegaHealthLimogesFrance
- Inserm, Bordeaux Population Health Research CenterUniversity of BordeauxBordeauxFrance
| | - Eliza Eleni‐Zacharoula Georgiou
- Department of PsychiatryPatras University General HospitalFaculty of Medicine, School of Health SciencesUniversity of PatrasPatrasGreece
| | - Ishtar Govia
- Caribbean Institute for Health ResearchThe University of the West Indies, JamaicaWest IndiesJamaica
- Institute for Global HealthUniversity College LondonLondonUK
| | - Lea T. Grinberg
- Department of Neurology and PathologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of PathologyUniversity of Sao PauloR. da Reitoria, R. Cidade UniversitáriaSão PauloSao PauloBrazil
| | - Maëlenn Guerchet
- Inserm U1094, IRD U270University of LimogesCHU Limoges, EpiMaCT ‐ Epidemiology of Chronic Diseases in Tropical ZoneInstitute of Epidemiology and Tropical NeurologyOmegaHealthLimogesFrance
| | - Seid Ali Gugssa
- Department of NeurologySchool of MedicineAddis Ababa UniversityAddis AbabaEthiopia
| | | | - Eef Hogervorst
- Loughborough UniversityLoughboroughUK
- Respati UniversityYogyakartaIndonesia
| | | | - Agustin Ibanez
- Latin American Institute for Brain Health (BrainLat)Universidad Adolfo IbanezPeñalolénSantiagoChile
- Global Brain Health Institute (GBHI)University California San Francisco (UCSF)San FranciscoCaliforniaUSA
- Global Brain Health Institute (GBHI)Trinity College DublinLloyd Building Trinity College DublinDublinIreland
- Cognitive Neuroscience Center (CNC)Universidad de San Andrés, and National Scientific and Technical Research Council (CONICET)VictoriaProvincia de Buenos AiresArgentina
| | - Masafumi Ihara
- Department of NeurologyNational Cerebral and Cardiovascular CenterSuitaOsakaJapan
| | - Thomas Gregor Issac
- Centre for Brain ResearchIndian Institute of Science (IISc)BengaluruKarnatakaIndia
| | - Linus Jönsson
- Department of NeurobiologyCare Science and Society, section for NeurogeriatricsKarolinska Institute, SolnavägenSolnaSweden
| | - Wambui M. Karanja
- Global Brain Health Institute (GBHI)Trinity College DublinLloyd Building Trinity College DublinDublinIreland
- Brain and Mind InstituteAga Khan UniversityNairobiKenya
| | - Joseph H. Lee
- Sergievsky CenterTaub Institute for Research on Alzheimer's Disease and the Aging BrainDepartments of Neurology and EpidemiologyColumbia UniversityNew YorkNew YorkUSA
| | - Iracema Leroi
- Global Brain Health Institute (GBHI)Trinity College DublinLloyd Building Trinity College DublinDublinIreland
| | | | - Facundo Francisco Manes
- Institute of Cognitive and Translational Neuroscience (INCYT)INECO FoundationFavaloro UniversityBuenos AiresArgentina
| | - Lingani Mbakile‐Mahlanza
- Global Brain Health Institute (GBHI)University California San Francisco (UCSF)San FranciscoCaliforniaUSA
- University of BotswanaGaboroneBotswana
| | - Bruce L. Miller
- Department of NeurologyMemory and Aging CenterUniversity of California San Francisco Weill Institute for NeurosciencesSan FranciscoCaliforniaUSA
| | | | - Victoria N. Mutiso
- Africa Mental Health Research and Training FoundationNairobiKenya
- Department of PsychiatryUniversity of NairobiNairobiKenya
- World Psychiatric Association Collaborating Centre for Research and TrainingNairobiKenya
| | | | - David M. Ndetei
- Africa Mental Health Research and Training FoundationNairobiKenya
- Department of PsychiatryUniversity of NairobiNairobiKenya
- World Psychiatric Association Collaborating Centre for Research and TrainingNairobiKenya
| | - Sam Nightingale
- Neuroscience InstituteUniversity of Cape TownCape TownSouth Africa
| | - Gabriela Novotni
- University Clinic of NeurologyMedical Faculty University Ss Cyril and Methodius Institute for Alzheimer's Disease and NeuroscienceSkopjeNorth Macedonia
| | - Primrose Nyamayaro
- Global Brain Health Institute (GBHI)Trinity College DublinLloyd Building Trinity College DublinDublinIreland
- Faculty of Medicine and Health SciencesUniversity of ZimbabweHarareZimbabwe
| | - Solomon Nyame
- Kintampo Health Research CentreGhana Health ServiceHospital RoadNear Kintampo‐north Municipal HospitalKintampoGhana
| | | | | | - Maira Okada de Oliveira
- Global Brain Health Institute (GBHI)University California San Francisco (UCSF)San FranciscoCaliforniaUSA
- Global Brain Health Institute (GBHI)Trinity College DublinLloyd Building Trinity College DublinDublinIreland
- Department of Psychiatry at Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Cognitive Neurology and Behavioral Unit (GNCC)University of Sao PauloR. da Reitoria, R. Cidade UniversitáriaSão PauloSao PauloBrazil
| | - Njideka U. Okubadejo
- Neurology UnitDepartment of MedicineFaculty of Clinical SciencesCollege of MedicineUniversity of LagosYabaLagosNigeria
| | - Martin Orrell
- Institute of Mental HealthUniversity of NottinghamNottinghamUK
| | - Stella‐Maria Paddick
- Newcastle UniversityNewcastle upon TyneUK
- Gateshead Health NHS Foundation TrustSheriff HillTyne and WearUK
| | - Margaret A. Pericak‐Vance
- John P Hussman Institute for Human GenomicsMiller School of MedicineUniversity of MiamiCoral GablesFloridaUSA
- Dr. John T Macdonald Foundation Department of Human GeneticsUniversity of Miami Miller School of MedicineCoral GablesFloridaUSA
| | - Zvezdan Pirtosek
- Faculty of MedicineUniversity Medical Centre LjubljanaLjubljanaSlovenia
| | - Felix Claude Victor Potocnik
- Old Age Psychiatry Unit, Depth PsychiatryStellenbosch UniversityWestern Cape, Stellenbosch CentralStellenboschSouth Africa
| | - Rema Raman
- Alzheimer's Therapeutic Research InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Mie Rizig
- Department of Neuromuscular DiseasesUCL Queen Square Institute of NeurologyQueen SquareLondonUK
| | - Mónica Rosselli
- Department of PsychologyCharles E. Schmidt College of ScienceFlorida Atlantic UniversityBoca RatonFloridaUSA
- Florida Alzheimer's Disease Research CenterGainesvilleFloridaUSA
| | | | - Claudia L. Satizabal
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Sciences CenterSan AntonioTexasUSA
- Department of NeurologyBoston University School of MedicineBostonMassachusettsUSA
- The Framingham Heart StudyFraminghamMassachusettsUSA
| | - Diego Sepulveda‐Falla
- Molecular Neuropathology of Alzheimer's DiseaseInstitute of NeuropathologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases and South Texas ADRCUT Health San AntonioSan AntonioTexasUSA
- University of Texas Health Sciences CenterSan AntonioTexasUSA
| | - Claire E. Sexton
- Division of Medical and Scientific RelationsAlzheimer's AssociationChicagoIllinoisUSA
| | - Ingmar Skoog
- Institute of Neuroscience and FysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Peter H. St George‐Hyslop
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainDepartment of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Cambridge Institute for Medical Research and Department of Clinical NeurosciencesSchool of Clinical MedicineUniversity of CambridgeAddenbrookes Biomedical CampusTrumpingtonCambridgeUK
- Department of Medicine (Neurology)Temerty Faculty of MedicineUniversity of Torontoand University Health Network27 King's College CirTorontoOntarioCanada
| | - Claudia Kimie Suemoto
- Division of GeriatricsUniversity of Sao Paulo Medical SchoolR. da Reitoria, R. Cidade UniversitáriaSão PauloSao PauloBrazil
| | - Prekshy Thapa
- Global Brain Health Institute (GBHI)Trinity College DublinLloyd Building Trinity College DublinDublinIreland
| | - Chinedu Theresa Udeh‐Momoh
- Global Brain Health Institute (GBHI)University California San Francisco (UCSF)San FranciscoCaliforniaUSA
- FINGERS Brain Health Institutec/o Stockholms SjukhemStockholmSweden
- Department of Epidemiology and PreventionWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
- Department of NeurobiologyCare Sciences and Society (NVS)Division of Clinical GeriatricsKarolinska Institute, SolnavägenSolnaSweden
- Imarisha Centre for Brain health and AgingBrain and Mind InstituteAga Khan UniversityNairobiKenya
| | - Victor Valcour
- Memory and Aging CenterDepartment of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Jeffery M. Vance
- John P Hussman Institute for Human GenomicsMiller School of MedicineUniversity of MiamiCoral GablesFloridaUSA
| | - Mathew Varghese
- St. John's Medical CollegeSarjapur ‐ Marathahalli Rd, beside Bank Of Baroda, John Nagar, KoramangalaBengaluruKarnatakaIndia
| | - Jaime H. Vera
- Department of Global Health and InfectionBrighton and Sussex Medical SchoolBrightonUK
| | - Richard W. Walker
- Population Health Sciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgGöteborgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyQueen Square, Queen SquareLondonUK
- UK Dementia Research Institute at UCLUniversity College LondonLondonUK
- Hong Kong Center for Neurodegenerative DiseasesClear Water BayHong KongChina
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Yared Z. Zewde
- Department of NeurologySchool of MedicineAddis Ababa UniversityAddis AbabaEthiopia
| | - Ozama Ismail
- Division of Medical and Scientific RelationsAlzheimer's AssociationChicagoIllinoisUSA
| |
Collapse
|
6
|
Soto-Mercado V, Mendivil-Perez M, Velez-Pardo C, Jimenez-Del-Rio M. Neuroprotective Effect of Combined Treatment with Epigallocatechin 3-Gallate and Melatonin on Familial Alzheimer's Disease PSEN1 E280A Cerebral Spheroids Derived from Menstrual Mesenchymal Stromal Cells. J Alzheimers Dis 2024; 99:S51-S66. [PMID: 36846998 DOI: 10.3233/jad-220903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Background Familial Alzheimer's disease (FAD) is caused by mutations in one or more of 3 genes known as AβPP, PSEN1, and PSEN2. There are currently no effective therapies for FAD. Hence, novel therapeutics are needed. Objective To analyze the effect of treatment with a combination of epigallocatechin-3-gallate (EGCG) and Melatonin (N-acetyl-5-methoxytryptamine, aMT) in a cerebral spheroid (CS) 3D in vitro model of PSEN 1 E280A FAD. Methods We developed a CS in vitro model based on menstrual stromal cells derived from wild-type (WT) and mutant PSEN1 E280A menstrual blood cultured in Fast-N-Spheres V2 medium. Results Beta-tubulin III, choline acetyltransferase, and GFAP in both WT and mutant CSs spontaneously expressed neuronal and astroglia markers when grown in Fast-N-Spheres V2 medium for 4 or 11 days. Mutant PSEN1 CSs had significantly increased levels of intracellular AβPP fragment peptides and concomitant appearance of oxidized DJ-1 as early as 4 days, and phosphorylated tau, decreased ΔΨm, and increased caspase-3 activity were observed on Day 11. Moreover, mutant CSs were unresponsive to acetylcholine. Treatment with a combination of EGCG and aMT decreased the levels of all typical pathological markers of FAD more efficiently than did EGCG or aMT alone, but aMT failed to restore Ca2+ influx in mutant CSs and decreased the beneficial effect of EGCG on Ca2+ influx in mutant CSs. Conclusion Treatment with a combination of EGCG and aMT can be of high therapeutic value due to the high antioxidant capacity and anti-amyloidogenic effect of both compounds.
Collapse
Affiliation(s)
- Viviana Soto-Mercado
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia, SIU Medellin, Colombia
| | - Miguel Mendivil-Perez
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia, SIU Medellin, Colombia
| | - Carlos Velez-Pardo
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia, SIU Medellin, Colombia
| | - Marlene Jimenez-Del-Rio
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia, SIU Medellin, Colombia
| |
Collapse
|
7
|
Gomez-Sequeda N, Mendivil-Perez M, Jimenez-Del-Rio M, Lopera F, Velez-Pardo C. Cholinergic-like neurons and cerebral spheroids bearing the PSEN1 p.Ile416Thr variant mirror Alzheimer's disease neuropathology. Sci Rep 2023; 13:12833. [PMID: 37553376 PMCID: PMC10409854 DOI: 10.1038/s41598-023-39630-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 07/27/2023] [Indexed: 08/10/2023] Open
Abstract
Familial Alzheimer's disease (FAD) is a complex neurodegenerative disorder for which there are no therapeutics to date. Several mutations in presenilin 1 (PSEN 1), which is the catalytic component of γ-secretase complex, are causal of FAD. Recently, the p.Ile416Thr (I416T) PSEN 1 mutation has been reported in large kindred in Colombia. However, cell and molecular information from I416T mutation is scarce. Here, we demonstrate that menstrual stromal cells (MenSCs)-derived planar (2D) PSEN 1 I416T cholinergic-like cells (ChLNS) and (3D) cerebral spheroids (CSs) reproduce the typical neuropathological markers of FAD in 4 post-transdifferentiating or 11 days of transdifferentiating, respectively. The models produce intracellular aggregation of APPβ fragments (at day 4 and 11) and phosphorylated protein TAU at residue Ser202/Thr205 (at day 11) suggesting that iAPPβ fragments precede p-TAU. Mutant ChLNs and CSs displayed DJ-1 Cys106-SO3 (sulfonic acid), failure of mitochondria membrane potential (ΔΨm), and activation of transcription factor c-JUN and p53, expression of pro-apoptotic protein PUMA, and activation of executer protein caspase 3 (CASP3), all markers of cell death by apoptosis. Moreover, we found that both mutant ChLNs and CSs produced high amounts of extracellular eAβ42. The I416T ChLNs and CSs were irresponsive to acetylcholine induced Ca2+ influx compared to WT. The I416T PSEN 1 mutation might work as dominant-negative PSEN1 mutation. These findings might help to understanding the recurring failures of clinical trials of anti-eAβ42, and support the view that FAD is triggered by the accumulation of other intracellular AβPP metabolites, rather than eAβ42.
Collapse
Affiliation(s)
- Nicolas Gomez-Sequeda
- Grupo de Neurociencias de Antioquia, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Torre 1, Laboratorio 412, Medellín, Colombia
| | - Miguel Mendivil-Perez
- Grupo de Neurociencias de Antioquia, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Torre 1, Laboratorio 412, Medellín, Colombia
| | - Marlene Jimenez-Del-Rio
- Grupo de Neurociencias de Antioquia, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Torre 1, Laboratorio 412, Medellín, Colombia
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Torre 1, Laboratorio 412, Medellín, Colombia
| | - Carlos Velez-Pardo
- Grupo de Neurociencias de Antioquia, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Torre 1, Laboratorio 412, Medellín, Colombia.
| |
Collapse
|
8
|
Mendivil-Perez M, Velez-Pardo C, Lopera F, Kosik KS, Jimenez-Del-Rio M. PSEN1 E280A Cholinergic-like Neurons and Cerebral Spheroids Derived from Mesenchymal Stromal Cells and from Induced Pluripotent Stem Cells Are Neuropathologically Equivalent. Int J Mol Sci 2023; 24:8957. [PMID: 37240306 PMCID: PMC10218810 DOI: 10.3390/ijms24108957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurological condition characterized by the severe loss of cholinergic neurons. Currently, the incomplete understanding of the loss of neurons has prevented curative treatments for familial AD (FAD). Therefore, modeling FAD in vitro is essential for studying cholinergic vulnerability. Moreover, to expedite the discovery of disease-modifying therapies that delay the onset and slow the progression of AD, we depend on trustworthy disease models. Although highly informative, induced pluripotent stem cell (iPSCs)-derived cholinergic neurons (ChNs) are time-consuming, not cost-effective, and labor-intensive. Other sources for AD modeling are urgently needed. Wild-type and presenilin (PSEN)1 p.E280A fibroblast-derived iPSCs, menstrual blood-derived menstrual stromal cells (MenSCs), and umbilical cord-derived Wharton Jelly's mesenchymal stromal cells (WJ-MSCs) were cultured in Cholinergic-N-Run and Fast-N-Spheres V2 medium to obtain WT and PSEN 1 E280A cholinergic-like neurons (ChLNs, 2D) and cerebroid spheroids (CSs, 3D), respectively, and to evaluate whether ChLNs/CSs can reproduce FAD pathology. We found that irrespective of tissue source, ChLNs/CSs successfully recapitulated the AD phenotype. PSEN 1 E280A ChLNs/CSs show accumulation of iAPPβ fragments, produce eAβ42, present TAU phosphorylation, display OS markers (e.g., oxDJ-1, p-JUN), show loss of ΔΨm, exhibit cell death markers (e.g., TP53, PUMA, CASP3), and demonstrate dysfunctional Ca2+ influx response to ACh stimuli. However, PSEN 1 E280A 2D and 3D cells derived from MenSCs and WJ-MSCs can reproduce FAD neuropathology more efficiently and faster (11 days) than ChLNs derived from mutant iPSCs (35 days). Mechanistically, MenSCs and WJ-MSCs are equivalent cell types to iPSCs for reproducing FAD in vitro.
Collapse
Affiliation(s)
- Miguel Mendivil-Perez
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, Calle 62#52-59, Building 1, Room 412, SIU, Medellin 050010, Colombia; (M.M.-P.); (C.V.-P.); (F.L.)
| | - Carlos Velez-Pardo
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, Calle 62#52-59, Building 1, Room 412, SIU, Medellin 050010, Colombia; (M.M.-P.); (C.V.-P.); (F.L.)
| | - Francisco Lopera
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, Calle 62#52-59, Building 1, Room 412, SIU, Medellin 050010, Colombia; (M.M.-P.); (C.V.-P.); (F.L.)
| | - Kenneth S. Kosik
- Neuroscience Research Institute, Department of Molecular Cellular Developmental Biology, University of California, Santa Barbara, CA 93106, USA;
| | - Marlene Jimenez-Del-Rio
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, Calle 62#52-59, Building 1, Room 412, SIU, Medellin 050010, Colombia; (M.M.-P.); (C.V.-P.); (F.L.)
| |
Collapse
|
9
|
Reiman EM, Pruzin JJ, Rios-Romenets S, Brown C, Giraldo M, Acosta-Baena N, Tobon C, Hu N, Chen Y, Ghisays V, Enos J, Goradia DD, Lee W, Luo J, Malek-Ahmadi M, Protas H, Thomas RG, Chen K, Su Y, Boker C, Mastroeni D, Alvarez S, Quiroz YT, Langbaum JB, Sink KM, Lopera F, Tariot PN. A public resource of baseline data from the Alzheimer's Prevention Initiative Autosomal-Dominant Alzheimer's Disease Trial. Alzheimers Dement 2023; 19:1938-1946. [PMID: 36373344 PMCID: PMC10262848 DOI: 10.1002/alz.12843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 09/01/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022]
Abstract
INTRODUCTION The Alzheimer's Prevention Initiative Autosomal-Dominant Alzheimer's Disease (API ADAD) Trial evaluated the anti-oligomeric amyloid beta (Aβ) antibody therapy crenezumab in cognitively unimpaired members of the Colombian presenilin 1 (PSEN1) E280A kindred. We report availability, methods employed to protect confidentiality and anonymity of participants, and process for requesting and accessing baseline data. METHODS We developed mechanisms to share baseline data from the API ADAD Trial in consultation with experts and other groups sharing data from Alzheimer's disease (AD) prevention trials, balancing the need to protect anonymity and trial integrity with making data broadly available to accelerate progress in the field. We pressure-tested deliberate and inadvertent potential threats under specific assumptions, employed a system to suppress or mask both direct and indirect identifying variables, limited and firewalled data managers, and put forth specific principles requisite to receive data. RESULTS Baseline demographic, PSEN1 E280A and apolipoprotein E genotypes, florbetapir and fluorodeoxyglucose positron emission tomography, magnetic resonance imaging, clinical, and cognitive data can now be requested by interested researchers. DISCUSSION Baseline data are publicly available; treatment data and biological samples, including baseline and treatment-related blood-based biomarker data will become available in accordance with our original trial agreement and subsequently developed Collaboration for Alzheimer's Prevention principles. Sharing of these data will allow exploration of important questions including the differential effects of initiating an investigational AD prevention therapy both before as well as after measurable Aβ plaque deposition.
Collapse
Affiliation(s)
- Eric M. Reiman
- Banner Alzheimer’s Institute, Phoenix, AZ, USA
- University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Jeremy J. Pruzin
- Banner Alzheimer’s Institute, Phoenix, AZ, USA
- University of Arizona College of Medicine, Phoenix, AZ, USA
| | | | - Chris Brown
- Banner Alzheimer’s Institute, Phoenix, AZ, USA
| | - Margarita Giraldo
- Grupo de Neurociencias de la Universidad de Antioquia, Medellin, Colombia
| | | | - Carlos Tobon
- Grupo de Neurociencias de la Universidad de Antioquia, Medellin, Colombia
| | - Nan Hu
- Genentech Inc., South San Francisco, CA, USA
| | | | | | | | | | - Wendy Lee
- Banner Alzheimer’s Institute, Phoenix, AZ, USA
| | - Ji Luo
- Banner Alzheimer’s Institute, Phoenix, AZ, USA
| | | | | | | | - Kewei Chen
- Banner Alzheimer’s Institute, Phoenix, AZ, USA
| | - Yi Su
- Banner Alzheimer’s Institute, Phoenix, AZ, USA
| | | | - Diego Mastroeni
- ASU-Banner Neurodegenerative Research Center, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | | | - Yakeel T. Quiroz
- Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA
| | - Jessica B. Langbaum
- Banner Alzheimer’s Institute, Phoenix, AZ, USA
- University of Arizona College of Medicine, Phoenix, AZ, USA
| | | | - Francisco Lopera
- Grupo de Neurociencias de la Universidad de Antioquia, Medellin, Colombia
| | - Pierre N. Tariot
- Banner Alzheimer’s Institute, Phoenix, AZ, USA
- University of Arizona College of Medicine, Phoenix, AZ, USA
| | | |
Collapse
|
10
|
Henao‐Restrepo J, López‐Murillo C, Valderrama‐Carmona P, Orozco‐Santa N, Gomez J, Gutiérrez‐Vargas J, Moraga R, Toledo J, Littau JL, Härtel S, Arboleda‐Velásquez JF, Sepulveda‐Falla D, Lopera F, Cardona‐Gómez GP, Villegas A, Posada‐Duque R. Gliovascular alterations in sporadic and familial Alzheimer's disease: APOE3 Christchurch homozygote glioprotection. Brain Pathol 2023; 33:e13119. [PMID: 36130084 PMCID: PMC10041169 DOI: 10.1111/bpa.13119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 08/30/2022] [Indexed: 11/28/2022] Open
Abstract
In response to brain insults, astrocytes become reactive, promoting protection and tissue repair. However, astroglial reactivity is typical of brain pathologies, including Alzheimer's disease (AD). Considering the heterogeneity of the reactive response, the role of astrocytes in the course of different forms of AD has been underestimated. Colombia has the largest human group known to have familial AD (FAD). This group carries the autosomal dominant and fully penetrant mutation E280A in PSEN1, which causes early-onset AD. Recently, our group identified an E280A carrier who did not develop FAD. The individual was homozygous for the Christchurch mutation R136S in APOE3 (APOEch). Remarkably, APOE is the main genetic risk factor for developing sporadic AD (SAD) and most of cerebral ApoE is produced by astroglia. Here, we characterized astrocyte properties related to reactivity, glutamate homeostasis, and structural integrity of the gliovascular unit (GVU), as factors that could underlie the pathogenesis or protection of AD. Specifically, through histological and 3D microscopy analyses of postmortem samples, we briefly describe the histopathology and cytoarchitecture of the frontal cortex of SAD, FAD, and APOEch, and demonstrate that, while astrodegeneration and vascular deterioration are prominent in SAD, FAD is characterized by hyperreactive-like glia, and APOEch displays the mildest astrocytic and vascular alterations despite having the highest burden of Aβ. Notably, astroglial, gliovascular, and vascular disturbances, as well as brain cell death, correlate with the specific astrocytic phenotypes identified in each condition. This study provides new insights into the potential relevance of the gliovasculature in the development and protection of AD. To our knowledge, this is the first study assessing the components of the GVU in human samples of SAD, FAD, and APOEch.
Collapse
Affiliation(s)
- Julián Henao‐Restrepo
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Carolina López‐Murillo
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Pablo Valderrama‐Carmona
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Natalia Orozco‐Santa
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Johana Gomez
- Grupo de Neurociencias de Antioquia, Facultad de MedicinaSIU, Universidad de AntioquiaMedellínColombia
| | - Johanna Gutiérrez‐Vargas
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Health Sciences FacultyRemington University CorporationMedellínColombia
| | - Renato Moraga
- Biomedical Neuroscience Institute BNI, Faculty of MedicineUniversity of ChileSantiagoChile
| | - Jorge Toledo
- Biomedical Neuroscience Institute BNI, Faculty of MedicineUniversity of ChileSantiagoChile
| | - Jessica Lisa Littau
- Molecular Neuropathology of Alzheimer's DiseaseInstitute of Neuropathology, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Steffen Härtel
- Biomedical Neuroscience Institute BNI, Faculty of MedicineUniversity of ChileSantiagoChile
| | - Joseph F. Arboleda‐Velásquez
- Schepens Eye Research Institute of Mass Eye and Ear, Department of OphthalmologyHarvard Medical SchoolBostonMassachusettsUSA
| | - Diego Sepulveda‐Falla
- Molecular Neuropathology of Alzheimer's DiseaseInstitute of Neuropathology, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Facultad de MedicinaSIU, Universidad de AntioquiaMedellínColombia
| | - Gloria Patricia Cardona‐Gómez
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Andrés Villegas
- Grupo de Neurociencias de Antioquia, Facultad de MedicinaSIU, Universidad de AntioquiaMedellínColombia
| | - Rafael Posada‐Duque
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| |
Collapse
|
11
|
Alvarez KLF, Aguilar-Pineda JA, Ortiz-Manrique MM, Paredes-Calderon MF, Cardenas-Quispe BC, Vera-Lopez KJ, Goyzueta-Mamani LD, Chavez-Fumagalli MA, Davila-Del-Carpio G, Peralta-Mestas A, Musolino PL, Lino Cardenas CL. Co-occurring pathogenic variants in 6q27 associated with dementia spectrum disorders in a Peruvian family. Front Mol Neurosci 2023; 16:1104585. [PMID: 36873109 PMCID: PMC9978490 DOI: 10.3389/fnmol.2023.1104585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/18/2023] [Indexed: 02/18/2023] Open
Abstract
Evidence suggests that there may be racial differences in risk factors associated with the development of Alzheimer's disease and related dementia (ADRD). We used whole-genome sequencing analysis and identified a novel combination of three pathogenic variants in the heterozygous state (UNC93A: rs7739897 and WDR27: rs61740334; rs3800544) in a Peruvian family with a strong clinical history of ADRD. Notably, the combination of these variants was present in two generations of affected individuals but absent in healthy members of the family. In silico and in vitro studies have provided insights into the pathogenicity of these variants. These studies predict that the loss of function of the mutant UNC93A and WDR27 proteins induced dramatic changes in the global transcriptomic signature of brain cells, including neurons, astrocytes, and especially pericytes and vascular smooth muscle cells, indicating that the combination of these three variants may affect the neurovascular unit. In addition, known key molecular pathways associated with dementia spectrum disorders were enriched in brain cells with low levels of UNC93A and WDR27. Our findings have thus identified a genetic risk factor for familial dementia in a Peruvian family with an Amerindian ancestral background.
Collapse
Affiliation(s)
- Karla Lucia F. Alvarez
- Laboratory of Genomics and Neurovascular Diseases, Universidad Católica de Santa María, Arequipa, Peru
| | | | | | | | - Bryan C. Cardenas-Quispe
- Division of Neurology, Psychiatry and Radiology of the National Hospital ESSALUD-HNCASE, Arequipa, Peru
| | - Karin Jannet Vera-Lopez
- Laboratory of Genomics and Neurovascular Diseases, Universidad Católica de Santa María, Arequipa, Peru
| | - Luis D. Goyzueta-Mamani
- Laboratory of Genomics and Neurovascular Diseases, Universidad Católica de Santa María, Arequipa, Peru
| | | | | | - Antero Peralta-Mestas
- Division of Neurology, Psychiatry and Radiology of the National Hospital ESSALUD-HNCASE, Arequipa, Peru
| | - Patricia L. Musolino
- Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
| | | |
Collapse
|
12
|
Reyes‐Dumeyer D, Faber K, Vardarajan B, Goate A, Renton A, Chao M, Boeve B, Cruchaga C, Pericak‐Vance M, Haines JL, Rosenberg R, Tsuang D, Sweet RA, Bennett DA, Wilson RS, Foroud T, Mayeux R. The National Institute on Aging Late-Onset Alzheimer's Disease Family Based Study: A resource for genetic discovery. Alzheimers Dement 2022; 18:1889-1897. [PMID: 34978149 PMCID: PMC9250549 DOI: 10.1002/alz.12514] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/25/2021] [Accepted: 08/11/2021] [Indexed: 02/02/2023]
Abstract
INTRODUCTION The National Institute on Aging Late-Onset Alzheimer's Disease Family Based Study (NIA-LOAD FBS) was established to study the genetic etiology of Alzheimer's disease (AD). METHODS Recruitment focused on families with two living affected siblings and a third first-degree relative similar in age with or without dementia. Uniform assessments were completed, DNA was obtained, as was neuropathology, when possible. Apolipoprotein E (APOE) genotypes, genome-wide single nucleotide polymorphism (SNP) arrays, and sequencing was completed in most families. RESULTS APOE genotype modified the age-at-onset in many large families. Novel variants and known variants associated with early- and late-onset AD and frontotemporal dementia were identified supporting an international effort to solve AD genetics. DISCUSSION The NIA-LOAD FBS is the largest collection of familial AD worldwide, and data or samples have been included in 123 publications addressing the genetic etiology of AD. Genetic heterogeneity and variability in the age-at-onset provides opportunities to investigate the complexity of familial AD.
Collapse
Affiliation(s)
- Dolly Reyes‐Dumeyer
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and the Aging Brain and the Gertrude H. Sergievsky Center, Columbia University in the City of New YorkNew YorkNew YorkUSA
| | - Kelley Faber
- Department of Medical and Molecular GeneticsNational Centralized Repository for Alzheimer's Disease and Related Dementias (NCRAD)Indiana University School of MedicineIndianapolisIndianaUSA
| | - Badri Vardarajan
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and the Aging Brain and the Gertrude H. Sergievsky Center, Columbia University in the City of New YorkNew YorkNew YorkUSA
| | - Alison Goate
- Department of Genetics & Genomic SciencesRonald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Alan Renton
- Department of Genetics & Genomic SciencesRonald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Michael Chao
- Department of Genetics & Genomic SciencesRonald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Brad Boeve
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
| | - Carlos Cruchaga
- Department of PsychiatryWashington University in St. LouisSt. LouisMissouriUSA
| | - Margaret Pericak‐Vance
- John P. Hussman Institute for Human GenomicsDr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of MedicineMiamiFloridaUSA
| | - Jonathan L. Haines
- Department of Population & Quantitative Health Sciences and Cleveland Institute for Computational BiologyCase Western Reserve UniversityClevelandOhioUSA
| | - Roger Rosenberg
- Department of NeurologyUniversity of Texas Southwestern Medical Center at DallasDallasTexasUSA
| | - Debby Tsuang
- GRECC VA Puget SoundDepartment of Psychiatry and Behavioral SciencesUniversity of WashingtonSeattleWashingtonUSA
| | - Robert A. Sweet
- Departments of Psychiatry and NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - David A. Bennett
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - Robert S. Wilson
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - Tatiana Foroud
- Department of Medical and Molecular GeneticsNational Centralized Repository for Alzheimer's Disease and Related Dementias (NCRAD)Indiana University School of MedicineIndianapolisIndianaUSA
| | - Richard Mayeux
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and the Aging Brain and the Gertrude H. Sergievsky Center, Columbia University in the City of New YorkNew YorkNew YorkUSA
| |
Collapse
|
13
|
Soto-Mercado V, Mendivil-Perez M, Velez-Pardo C, Jimenez-Del-Rio M. (-)-Epigallocatechin-3-Gallate Diminishes Intra-and Extracellular Amyloid-Induced Cytotoxic Effects on Cholinergic-like Neurons from Familial Alzheimer's Disease PSEN1 E280A. Biomolecules 2021; 11:biom11121845. [PMID: 34944489 PMCID: PMC8699501 DOI: 10.3390/biom11121845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/26/2021] [Accepted: 11/26/2021] [Indexed: 12/26/2022] Open
Abstract
Alzheimer’s disease (AD) is a complex neurodegenerative disease characterized by functional disruption, death of cholinergic neurons (ChNs) because of intracellular and extracellular Aβ aggregates, and hyperphosphorylation of protein TAU (p-TAU). To date, there are no efficient therapies against AD. Therefore, new therapies for its treatment are in need. The goal of this investigation was to evaluate the effect of the polyphenol epigallocatechin-3-gallate (EGCG) on cholinergic-like neurons (ChLNs) bearing the mutation E280A in PRESENILIN 1 (PSEN1 E280A). To this aim, wild-type (WT) and PSEN1 E280A ChLNs were exposed to EGCG (5–50 μM) for 4 days. Untreated or treated neurons were assessed for biochemical and functional analysis. We found that EGCG (50 μM) significantly inhibited the aggregation of (i)sAPPβf, blocked p-TAU, increased ∆Ψm, decreased oxidation of DJ-1 at residue Cys106-SH, and inhibited the activation of transcription factor c-JUN and P53, PUMA, and CASPASE-3 in mutant ChLNs compared to WT. Although EGCG did not reduce (e)Aβ42, the polyphenol reversed Ca2+ influx dysregulation as a response to acetylcholine (ACh) stimuli in PSEN1 E280A ChLNs, inhibited the activation of transcription factor NF-κB, and reduced the secretion of pro-inflammatory IL-6 in wild-type astrocyte-like cells (ALCs) when exposed to mutant ChLNs culture supernatant. Taken together, our findings suggest that the EGCG might be a promising therapeutic approach for the treatment of FAD.
Collapse
|
14
|
Hampel H, Hardy J, Blennow K, Chen C, Perry G, Kim SH, Villemagne VL, Aisen P, Vendruscolo M, Iwatsubo T, Masters CL, Cho M, Lannfelt L, Cummings JL, Vergallo A. The Amyloid-β Pathway in Alzheimer's Disease. Mol Psychiatry 2021; 26:5481-5503. [PMID: 34456336 PMCID: PMC8758495 DOI: 10.1038/s41380-021-01249-0] [Citation(s) in RCA: 595] [Impact Index Per Article: 198.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/19/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023]
Abstract
Breakthroughs in molecular medicine have positioned the amyloid-β (Aβ) pathway at the center of Alzheimer's disease (AD) pathophysiology. While the detailed molecular mechanisms of the pathway and the spatial-temporal dynamics leading to synaptic failure, neurodegeneration, and clinical onset are still under intense investigation, the established biochemical alterations of the Aβ cycle remain the core biological hallmark of AD and are promising targets for the development of disease-modifying therapies. Here, we systematically review and update the vast state-of-the-art literature of Aβ science with evidence from basic research studies to human genetic and multi-modal biomarker investigations, which supports a crucial role of Aβ pathway dyshomeostasis in AD pathophysiological dynamics. We discuss the evidence highlighting a differentiated interaction of distinct Aβ species with other AD-related biological mechanisms, such as tau-mediated, neuroimmune and inflammatory changes, as well as a neurochemical imbalance. Through the lens of the latest development of multimodal in vivo biomarkers of AD, this cross-disciplinary review examines the compelling hypothesis- and data-driven rationale for Aβ-targeting therapeutic strategies in development for the early treatment of AD.
Collapse
Affiliation(s)
- Harald Hampel
- Eisai Inc., Neurology Business Group, Woodcliff Lake, NJ, USA.
| | - John Hardy
- UK Dementia Research Institute at UCL and Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Christopher Chen
- Memory Aging and Cognition Centre, Departments of Pharmacology and Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - George Perry
- Department of Biology and Neurosciences Institute, University of Texas at San Antonio (UTSA), San Antonio, TX, USA
| | - Seung Hyun Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea; Cell Therapy Center, Hanyang University Hospital, Seoul, Republic of Korea
| | - Victor L Villemagne
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia
| | - Paul Aisen
- USC Alzheimer's Therapeutic Research Institute, San Diego, CA, USA
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Colin L Masters
- Laureate Professor of Dementia Research, Florey Institute and The University of Melbourne, Parkville, VIC, Australia
| | - Min Cho
- Eisai Inc., Neurology Business Group, Woodcliff Lake, NJ, USA
| | - Lars Lannfelt
- Uppsala University, Department of of Public Health/Geriatrics, Uppsala, Sweden
- BioArctic AB, Stockholm, Sweden
| | - Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Andrea Vergallo
- Eisai Inc., Neurology Business Group, Woodcliff Lake, NJ, USA.
| |
Collapse
|
15
|
Llibre-Guerra JJ, Li Y, Allegri RF, Mendez PC, Surace EI, Llibre-Rodriguez JJ, Sosa AL, Aláez-Verson C, Longoria EM, Tellez A, Carrillo-Sánchez K, Flores-Lagunes LL, Sánchez V, Takada LT, Nitrini R, Ferreira-Frota NA, Benevides-Lima J, Lopera F, Ramírez L, Jiménez-Velázquez I, Schenk C, Acosta D, Behrens MI, Doering M, Ziegemeier E, Morris JC, McDade E, Bateman RJ. Dominantly inherited Alzheimer's disease in Latin America: Genetic heterogeneity and clinical phenotypes. Alzheimers Dement 2021; 17:653-664. [PMID: 33226734 PMCID: PMC8140610 DOI: 10.1002/alz.12227] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/08/2020] [Accepted: 10/09/2020] [Indexed: 01/21/2023]
Abstract
INTRODUCTION A growing number of dominantly inherited Alzheimer's disease (DIAD) cases have become known in Latin American (LatAm) in recent years. However, questions regarding mutation distribution and frequency by country remain open. METHODS A literature review was completed aimed to provide estimates for DIAD pathogenic variants in the LatAm population. The search strategies were established using a combination of standardized terms for DIAD and LatAm. RESULTS Twenty-four DIAD pathogenic variants have been reported in LatAm countries. Our combined dataset included 3583 individuals at risk; countries with highest DIAD frequencies were Colombia (n = 1905), Puerto Rico (n = 672), and Mexico (n = 463), usually attributable to founder effects. We found relatively few reports with extensive documentation on biomarker profiles and disease progression. DISCUSSION Future DIAD studies will be required in LatAm, albeit with a more systematic approach to include fluid biomarker and imaging studies. Regional efforts are under way to extend the DIAD observational studies and clinical trials to Latin America.
Collapse
Affiliation(s)
- Jorge J Llibre-Guerra
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yan Li
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Ricardo F Allegri
- Department of Cognitive Neurology, Institute for Neurological Research Fleni, Buenos Aires, Argentina
| | - Patricio Chrem Mendez
- Department of Cognitive Neurology, Institute for Neurological Research Fleni, Buenos Aires, Argentina
| | - Ezequiel I Surace
- Department of Cognitive Neurology, Institute for Neurological Research Fleni, Buenos Aires, Argentina
| | | | - Ana Luisa Sosa
- Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico, Mexico City, Mexico
| | - Carmen Aláez-Verson
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica, Ciudad de México, México
| | | | - Alberto Tellez
- Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico, Mexico City, Mexico
| | - Karol Carrillo-Sánchez
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica, Ciudad de México, México
| | | | - Victor Sánchez
- Department of Neurology, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, Brazil
| | | | | | | | | | - Francisco Lopera
- University of Puerto Rico School of Medicine, San Juan, Puerto Rico, USA
| | - Laura Ramírez
- University of Puerto Rico School of Medicine, San Juan, Puerto Rico, USA
| | | | - Christian Schenk
- Universidad Nacional Pedro Henríquez Ureña, Santo Domingo, Republica Dominicana
| | - Daisy Acosta
- Departamento de Neurología y Neurocirugía Hospital Clínico, Departamento de Neurociencias, Centro de Investigación Clínica Avanzada (CICA), Universidad de Chile & Clínica Alemana, Santiago, Chile
| | - María Isabel Behrens
- Becker Medical Library, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michelle Doering
- Department of Biostatistics, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Ellen Ziegemeier
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - John C Morris
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Eric McDade
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Randall J Bateman
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
16
|
Pérez-López N, Martín C, García B, Solís-Hernández MP, Rodríguez D, Alcalde I, Merayo J, Fernández-Vega I, Quirós LM. Alterations in the Expression of the Genes Responsible for the Synthesis of Heparan Sulfate in Brains With Alzheimer Disease. J Neuropathol Exp Neurol 2021; 80:446-456. [PMID: 33779723 DOI: 10.1093/jnen/nlab028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The saccharide chains of heparan sulfate appear to be involved in several aspects Alzheimer disease (AD) pathogenesis. Their structural complexity is due to the expression of different isoenzymes. We studied the differential transcription of heparan sulfate chain biosynthesis in AD brains, analyzing different brain regions in patients with different extents of AD pathology. The transcriptomic study was performed by RT-PCR using samples of amygdala, anterior hippocampus, posterior hippocampus, claustrum, calcarine fissure, globus pallidus and cerebellum from patients with mild, moderate, or severe AD, as well as healthy individuals. Certain heparan sulfate epitopes were also detected by immunohistochemistry. Several genes, across all stages of heparan sulfate synthesis, showed altered transcription in different brain regions of AD patients. The numbers of alterations were greater in in moderate versus mild AD patients. In severe patients, there were fewer alterations in genes related to early stages of biosynthesis, and overexpression of genes involved in late stages. The alterations correlated with progressive brain atrophy, although alterations were more common in the cerebellum. Detection of some heparan sulfate epitopes by immunohistochemistry was consistent with previous studies. In conclusion, transcriptional alterations in the biosynthetic genes of heparan sulfate depend on the brain region and the degree of AD pathology.
Collapse
Affiliation(s)
- Natalia Pérez-López
- From the Instituto Universitario Fernández-Vega, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Carla Martín
- From the Instituto Universitario Fernández-Vega, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,Department of Functional Biology, University of Oviedo, Oviedo, Spain
| | - Beatriz García
- Department of Functional Biology, University of Oviedo, Oviedo, Spain
| | | | - David Rodríguez
- Department of Biochemistry and Molecular Biology, University of Oviedo, Oviedo, Spain.,Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, Oviedo, Spain
| | - Ignacio Alcalde
- From the Instituto Universitario Fernández-Vega, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Jesús Merayo
- From the Instituto Universitario Fernández-Vega, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,Department of Surgery and Medical-surgical Specialties, University of Oviedo, Oviedo, Spain
| | - Iván Fernández-Vega
- From the Instituto Universitario Fernández-Vega, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,Department of Surgery and Medical-surgical Specialties, University of Oviedo, Oviedo, Spain.,Department of Pathology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Luis M Quirós
- From the Instituto Universitario Fernández-Vega, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,Department of Functional Biology, University of Oviedo, Oviedo, Spain
| |
Collapse
|
17
|
Soto-Mercado V, Mendivil-Perez M, Jimenez-Del-Rio M, Velez-Pardo C. Multi-Target Effects of the Cannabinoid CP55940 on Familial Alzheimer's Disease PSEN1 E280A Cholinergic-Like Neurons: Role of CB1 Receptor. J Alzheimers Dis 2020; 82:S359-S378. [PMID: 33252082 PMCID: PMC8293648 DOI: 10.3233/jad-201045] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by structural damage, death, and functional disruption of cholinergic neurons (ChNs) as a result of intracellular amyloid-β (Aβ) aggregation, extracellular neuritic plaques, and hyperphosphorylation of protein tau (p-Tau) overtime. OBJECTIVE To evaluate the effect of the synthetic cannabinoid CP55940 (CP) on PSEN1 E280A cholinergic-like nerve cells (PSEN1 ChLNs)-a natural model of familial AD. METHODS Wild type (WT) and PSEN1 ChLNs were exposed to CP (1μM) only or in the presence of the CB1 and CB2 receptors (CB1Rs, CB2Rs) inverse agonist SR141716 (1μM) and SR144528 (1μM) respectively, for 24 h. Untreated or treated neurons were assessed for biochemical and functional analysis. RESULTS CP in the presence of both inverse agonists (hereafter SR) almost completely inhibits the aggregation of intracellular sAβPPβf and p-Tau, increases ΔΨm, decreases oxidation of DJ-1Cys106-SH residue, and blocks the activation of c-Jun, p53, PUMA, and caspase-3 independently of CB1Rs signaling in mutant ChLNs. CP also inhibits the generation of reactive oxygen species partially dependent on CB1Rs. Although CP reduced extracellular Aβ42, it was unable to reverse the Ca2+ influx dysregulation as a response to acetylcholine stimuli in mutant ChLNs. Exposure to anti-Aβ antibody 6E10 (1:300) in the absence or presence of SR plus CP completely recovered transient [Ca2+]i signal as a response to acetylcholine in mutant ChLNs. CONCLUSION Taken together our findings suggest that the combination of cannabinoids, CB1Rs inverse agonists, and anti-Aβ antibodies might be a promising therapeutic approach for the treatment of familial AD.
Collapse
Affiliation(s)
- Viviana Soto-Mercado
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), SIU Medellin, Colombia
| | - Miguel Mendivil-Perez
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), SIU Medellin, Colombia
| | - Marlene Jimenez-Del-Rio
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), SIU Medellin, Colombia
| | - Carlos Velez-Pardo
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), SIU Medellin, Colombia
| |
Collapse
|
18
|
Tuazon AMDA, Lott P, Bohórquez M, Benavides J, Ramirez C, Criollo A, Estrada-Florez A, Mateus G, Velez A, Carmona J, Olaya J, Garcia E, Polanco-Echeverry G, Stultz J, Alvarez C, Tapia T, Ashton-Prolla P, Vega A, Lazaro C, Tornero E, Martinez-Bouzas C, Infante M, De La Hoya M, Diez O, Browning BL, Rannala B, Teixeira MR, Carvallo P, Echeverry M, Carvajal-Carmona LG. Haplotype analysis of the internationally distributed BRCA1 c.3331_3334delCAAG founder mutation reveals a common ancestral origin in Iberia. Breast Cancer Res 2020; 22:108. [PMID: 33087180 PMCID: PMC7579869 DOI: 10.1186/s13058-020-01341-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/16/2020] [Indexed: 12/02/2022] Open
Abstract
Background The BRCA1 c.3331_3334delCAAG founder mutation has been reported in hereditary breast and ovarian cancer families from multiple Hispanic groups. We aimed to evaluate BRCA1 c.3331_3334delCAAG haplotype diversity in cases of European, African, and Latin American ancestry. Methods BC mutation carrier cases from Colombia (n = 32), Spain (n = 13), Portugal (n = 2), Chile (n = 10), Africa (n = 1), and Brazil (n = 2) were genotyped with the genome-wide single nucleotide polymorphism (SNP) arrays to evaluate haplotype diversity around BRCA1 c.3331_3334delCAAG. Additional Portuguese (n = 13) and Brazilian (n = 18) BC mutation carriers were genotyped for 15 informative SNPs surrounding BRCA1. Data were phased using SHAPEIT2, and identical by descent regions were determined using BEAGLE and GERMLINE. DMLE+ was used to date the mutation in Colombia and Iberia. Results The haplotype reconstruction revealed a shared 264.4-kb region among carriers from all six countries. The estimated mutation age was ~ 100 generations in Iberia and that it was introduced to South America early during the European colonization period. Conclusions Our results suggest that this mutation originated in Iberia and later introduced to Colombia and South America at the time of Spanish colonization during the early 1500s. We also found that the Colombian mutation carriers had higher European ancestry, at the BRCA1 gene harboring chromosome 17, than controls, which further supported the European origin of the mutation. Understanding founder mutations in diverse populations has implications in implementing cost-effective, ancestry-informed screening.
Collapse
Affiliation(s)
| | - Paul Lott
- Genome Center, University of California Davis, Davis, CA, USA
| | | | | | | | | | | | | | - Alejandro Velez
- Hospital Pablo Tobon Uribe, Medellín, Colombia.,Dinamica IPS, Medellín, Colombia
| | | | - Justo Olaya
- Hospital Universitario Hernando Moncaleano Perdomo, Neiva, Colombia
| | - Elisha Garcia
- Genome Center, University of California Davis, Davis, CA, USA
| | | | - Jacob Stultz
- Genome Center, University of California Davis, Davis, CA, USA
| | | | - Teresa Tapia
- Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patricia Ashton-Prolla
- Department of Genetics, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Post-graduate Course in Genetics and Molecular Biology, UFRGS, Porto Alegre, Brazil.,Medical Genetics Service, Hospital de Clinicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | | | - Ana Vega
- Fundación Pública Galega de Medicina Xenómica, Grupo de Medicina Xenómica-USC, CIBERER, IDIS, Santiago de Compostela, Spain
| | - Conxi Lazaro
- Hereditary Cancer Program, Catalan Institute of Oncology, Oncobell Program-IDIBELL, Hospitalet de Llobregat, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Eva Tornero
- Hereditary Cancer Program, Catalan Institute of Oncology, Oncobell Program-IDIBELL, Hospitalet de Llobregat, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | | | - Mar Infante
- Cancer Genetics Group, Institute of Genetics and Molecular Biology (UVa-CSIC), Valladolid, Spain
| | - Miguel De La Hoya
- Laboratorio de Oncología Molecular, Hospital Clínico San Carlos. IdISSC (Instituto de Investigación Sanitaria San Carlos), Madrid, Spain
| | - Orland Diez
- Grupo de Cáncer Hereditario, Instituto Oncológico Vall d'Hebron (VHIO), Madrid, Spain
| | - Brian L Browning
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA
| | | | - Bruce Rannala
- Department of Evolution and Ecology, University of California Davis, Davis, CA, USA
| | - Manuel R Teixeira
- Portuguese Oncology Institute of Porto (IPO Porto) and Biomedical Sciences Institute (ICBAS), University of Porto, Porto, Portugal
| | - Pilar Carvallo
- Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Luis G Carvajal-Carmona
- Genome Center, University of California Davis, Davis, CA, USA. .,Division de Investigaciones, Fundacion de Genética y Genómica, Ibague, Colombia. .,University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA. .,Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA.
| |
Collapse
|
19
|
Abstract
Alzheimer’s disease (AD) and frontotemporal dementia (FTD) are neurodegenerative
disorders that result in a significant burden to both patients and caregivers.
By 2050, the number of people with dementia in Latin America will increase
4-fold. A deep understanding of the relevant genetic factors of AD and FTD is
fundamental to tackle this reality through prevention. A review of different
genetic variants that cause AD or FTD in Latin America was conducted. We
searched Medline and PubMed databases using the keywords “Alzheimer’s disease,”
“frontotemporal dementia,” “mutation,” “America,” and “Latin America,” besides
specific Latin American countries. Forty-five items were chosen and analyzed.
PSEN1 mutations are the commonest cause of genetic
early-onset Alzheimer’s disease (EOAD), followed by PSEN2 and
APP mutations. Genetic FTD can be mainly explained by
GRN and MAPT mutations, as well as
C9orf72 G4C2 repeat expansion. APOE ε4 can
modify the prevalence and incidence of late-onset Alzheimer’s disease (LOAD), in
addition to the cognitive performance in affected carriers.
Collapse
Affiliation(s)
- Claudia Ramos
- Neurosciences Group of Antioquia, School of Medicine, Universidad de Antioquia - Medellín, Colombia
| | - David Aguillon
- Neurosciences Group of Antioquia, School of Medicine, Universidad de Antioquia - Medellín, Colombia
| | - Christian Cordano
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco - San Francisco, CA, United States
| | - Francisco Lopera
- Neurosciences Group of Antioquia, School of Medicine, Universidad de Antioquia - Medellín, Colombia
| |
Collapse
|
20
|
Soto-Mercado V, Mendivil-Perez M, Velez-Pardo C, Lopera F, Jimenez-Del-Rio M. Cholinergic-like neurons carrying PSEN1 E280A mutation from familial Alzheimer's disease reveal intraneuronal sAPPβ fragments accumulation, hyperphosphorylation of TAU, oxidative stress, apoptosis and Ca2+ dysregulation: Therapeutic implications. PLoS One 2020; 15:e0221669. [PMID: 32437347 PMCID: PMC7241743 DOI: 10.1371/journal.pone.0221669] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 05/06/2020] [Indexed: 01/31/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by progressive memory loss and cognitive disturbance as a consequence of the loss of cholinergic neurons in the brain, neuritic plaques and hyperphosphorylation of TAU protein. Although the underlying mechanisms leading to these events are unclear, mutations in presenilin 1 (PSEN1), e.g., E280A (PSEN1 E280A), are causative factors for autosomal dominant early-onset familial AD (FAD). Despite advances in the understanding of the physiopathology of AD, there are no efficient therapies to date. Limitations in culturing brain-derived live neurons might explain the limited effectiveness of AD research. Here, we show that mesenchymal stromal (stem) cells (MSCs) can be used to model FAD, providing novel opportunities to study cellular mechanisms and to establish therapeutic strategies. Indeed, we cultured MSCs with the FAD mutation PSEN1 E280A and wild-type (WT) PSEN1 from umbilical cords and characterized the transdifferentiation of these cells into cholinergic-like neurons (ChLNs). PSEN1 E280A ChLNs but not WT PSEN1 ChLNs exhibited increased intracellular soluble amyloid precursor protein (sAPPf) fragments and extracellular Aβ42 peptide and TAU phosphorylation (at residues Ser202/Thr205), recapitulating the molecular pathogenesis of FAD caused by mutant PSEN1. Furthermore, PSEN1 E280A ChLNs presented oxidative stress (OS) as evidenced by the oxidation of DJ-1Cys106-SH into DJ-1Cys106-SO3 and the detection of DCF-positive cells and apoptosis markers such as activated pro-apoptosis proteins p53, c-JUN, PUMA and CASPASE-3 and the concomitant loss of the mitochondrial membrane potential and DNA fragmentation. Additionally, mutant ChLNs displayed Ca2+ flux dysregulation and deficient acetylcholinesterase (AChE) activity compared to control ChLNs. Interestingly, the inhibitor JNK SP600125 almost completely blocked TAU phosphorylation. Our findings demonstrate that FAD MSC-derived cholinergic neurons with the PSEN1 E280A mutation provide important clues for the identification of targetable pathological molecules.
Collapse
Affiliation(s)
- Viviana Soto-Mercado
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), SIU Medellin, Medellin, Colombia
| | - Miguel Mendivil-Perez
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), SIU Medellin, Medellin, Colombia
| | - Carlos Velez-Pardo
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), SIU Medellin, Medellin, Colombia
| | - Francisco Lopera
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), SIU Medellin, Medellin, Colombia
| | - Marlene Jimenez-Del-Rio
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), SIU Medellin, Medellin, Colombia
- * E-mail:
| |
Collapse
|
21
|
Rios-Romenets S, Lopera F, Sink KM, Hu N, Lian Q, Guthrie H, Smith J, Cho W, Mackey H, Langbaum JB, Thomas RG, Giraldo-Chica M, Tobon C, Acosta-Baena N, Muñoz C, Ospina P, Tirado V, Henao E, Bocanegra Y, Chen K, Su Y, Goradia D, Thiyyagura P, VanGilder PS, Luo J, Ghisays V, Lee W, Malek-Ahmadi MH, Protas HD, Chen Y, Quiroz YT, Reiman EM, Tariot PN. Baseline demographic, clinical, and cognitive characteristics of the Alzheimer's Prevention Initiative (API) Autosomal-Dominant Alzheimer's Disease Colombia Trial. Alzheimers Dement 2020; 16:1023-1030. [PMID: 32418361 PMCID: PMC7819133 DOI: 10.1002/alz.12109] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 01/26/2020] [Accepted: 02/21/2020] [Indexed: 01/22/2023]
Abstract
INTRODUCTION The API AutosomalDominant AD (ADAD) Colombia Trial is a placebo-controlled clinical trial of crenezumab in 252 cognitively unimpaired 30 to 60-year-old Presenilin 1 (PSEN1) E280A kindred members, including mutation carriers randomized to active treatment or placebo and non-carriers who receive placebo. METHODS Of the 252 enrolled, we present data on a total of 242 mutation carriers and non-carriers matched by age range, excluding data on 10 participants to protect participant confidentiality, genetic status, and trial integrity. RESULTS We summarize demographic, clinical, cognitive, and behavioral data from 167 mutation carriers and 75 non-carriers, 30 to 53 years of age. Carriers were significantly younger than non-carriers ((mean age ± SD) 37 ± 5 vs 42 ± 6), had significantly lower Mini Mental Status Exam (MMSE) scores (28.8 ± 1.4 vs 29.2 ± 1.0), and had consistently lower memory scores. DISCUSSION Although PSEN1 E280A mutation carriers in the Trial are cognitively unimpaired, they have slightly lower MMSE and memory scores than non-carriers. Their demographic characteristics are representative of the local population.
Collapse
Affiliation(s)
| | - Francisco Lopera
- Neurosciences Group of Antioquia/University of Antioquia, Medellin, Colombia
| | - Kaycee M Sink
- Genentech Inc., South San Francisco, California, USA
| | - Nan Hu
- Genentech Inc., South San Francisco, California, USA
| | - Qinshu Lian
- Genentech Inc., South San Francisco, California, USA
| | | | | | - William Cho
- Genentech Inc., South San Francisco, California, USA
| | - Howard Mackey
- Genentech Inc., South San Francisco, California, USA
| | | | | | | | - Carlos Tobon
- Neurosciences Group of Antioquia/University of Antioquia, Medellin, Colombia
| | | | - Claudia Muñoz
- Neurosciences Group of Antioquia/University of Antioquia, Medellin, Colombia
| | - Paula Ospina
- Neurosciences Group of Antioquia/University of Antioquia, Medellin, Colombia
| | - Victoria Tirado
- Neurosciences Group of Antioquia/University of Antioquia, Medellin, Colombia
| | - Eliana Henao
- Neurosciences Group of Antioquia/University of Antioquia, Medellin, Colombia
| | - Yamile Bocanegra
- Neurosciences Group of Antioquia/University of Antioquia, Medellin, Colombia
| | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, Arizona, USA
| | - Yi Su
- Banner Alzheimer's Institute, Phoenix, Arizona, USA
| | | | | | | | - Ji Luo
- Banner Alzheimer's Institute, Phoenix, Arizona, USA
| | | | - Wendy Lee
- Banner Alzheimer's Institute, Phoenix, Arizona, USA
| | | | | | - Yinghua Chen
- Banner Alzheimer's Institute, Phoenix, Arizona, USA
| | - Yakeel T Quiroz
- Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | -
- Neurosciences Group of Antioquia/University of Antioquia, Medellin, Colombia
| |
Collapse
|
22
|
Withers M, Sayegh P, Rodriguez-Agudelo Y, Ernstrom K, Raman R, Montoya L, Zuno-Reyes A, Mosieri C, Matute E, Ringman JM. A mixed-methods study of cultural beliefs about dementia and genetic testing among Mexicans and Mexican-Americans at-risk for autosomal dominant Alzheimer's disease. J Genet Couns 2019; 28:921-932. [PMID: 31207006 PMCID: PMC7500864 DOI: 10.1002/jgc4.1133] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 04/30/2019] [Accepted: 05/06/2019] [Indexed: 01/04/2023]
Abstract
Trials to prevent autosomal dominantly inherited Alzheimer's disease (ADAD) are critical and timely. However, cultural beliefs about AD and genetic testing may preclude informed consent and participation, especially among racial/ethnic minorities. This mixed-methods study examines cultural beliefs about AD and genetic screening among at-risk populations of Mexican heritage. We surveyed 86 Mexican and 37 Mexican-American family members of patients with ADAD and interviewed 18 respondents in Mexico to explore perceptions and knowledge regarding AD and genetic testing. While most respondents understood that AD is inherited in their families, they also had limited understanding of the genetic mechanisms behind AD. Many believed that AD is a normal part of aging or that it is a mental illness caused by bad habits. However, beliefs that AD is caused by a curse or God's will were uncommon. The interviews demonstrated that very few at-risk respondents understood their own risk for harboring the mutation causing AD in their family. Once informed, most expressed a strong interest in genetic testing, largely motivated by the desire to be better prepared for the development of AD. Health professionals treating and investigators enrolling members from families with ADAD cannot assume that they fully understand the nature of the illness; therefore, providers should provide comprehensive information about ADAD and genetic testing.
Collapse
Affiliation(s)
- Mellissa Withers
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Philip Sayegh
- Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA
| | | | - Karin Ernstrom
- Alzheimer’s Therapeutic Research Institute, University of Southern California, Los Angeles, California, USA
| | - Rema Raman
- Alzheimer’s Therapeutic Research Institute, University of Southern California, Los Angeles, California, USA
| | - Lucy Montoya
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| | | | - Chizoba Mosieri
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Esmeralda Matute
- Institute of Neurosciences, University of Guadalajara, Guadalajara, Mexico
| | - John M. Ringman
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
23
|
Homozygosity for the A431E mutation in PSEN1 presenting with a relatively aggressive phenotype. Neurosci Lett 2019; 699:195-198. [PMID: 30716424 DOI: 10.1016/j.neulet.2019.01.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/20/2019] [Accepted: 01/28/2019] [Indexed: 11/22/2022]
Abstract
OBJECTIVE We report a 35 year-old male with childhood learning disability and early onset dementia who is homozygous for the A431E variant in the PSEN1 gene. Presenilin1 mutations are associated with autosomal dominant Alzheimer's dementia with young and somewhat stereotyped onset. Such variants may cause Alzheimer's dementia through aberrant processing of amyloid precursor protein through effects on γ-secretase activity. γ-secretase is involved in the cleavage of many proteins critical to normal function, including brain development. Therefore, manifestations in persons without normal Presenilin1 function is of interest. METHODS Clinical evaluation including family history, examination, brain MRI, and genetic analysis. RESULTS Our patient had mild developmental delay, chronic nighttime behavioral disturbance, and onset of progressive cognitive deficits at age 33. Clinical evaluation demonstrated spastic paraparesis and pseudobulbar affect. Brain MRI revealed cerebral atrophy disproportionate to age. Chronic microhemorrhages within bilateral occipital, temporal, and right frontal lobes were seen. Sanger sequencing confirmed homozygosity for the A431E variant in PSEN1, which is a known pathogenic variant causing autosomal dominant Alzheimer's dementia. CONCLUSIONS Our report demonstrates that homozygosity for pathogenic Presenilin1 variants is compatible with life, though may cause a more aggressive phenotype with younger age of onset and possibly REM behavior disorder.
Collapse
|
24
|
Jacobs HIL, Hopkins DA, Mayrhofer HC, Bruner E, van Leeuwen FW, Raaijmakers W, Schmahmann JD. The cerebellum in Alzheimer's disease: evaluating its role in cognitive decline. Brain 2019; 141:37-47. [PMID: 29053771 DOI: 10.1093/brain/awx194] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 06/12/2017] [Indexed: 12/12/2022] Open
Abstract
The cerebellum has long been regarded as essential only for the coordination of voluntary motor activity and motor learning. Anatomical, clinical and neuroimaging studies have led to a paradigm shift in the understanding of the cerebellar role in nervous system function, demonstrating that the cerebellum appears integral also to the modulation of cognition and emotion. The search to understand the cerebellar contribution to cognitive processing has increased interest in exploring the role of the cerebellum in neurodegenerative and neuropsychiatric disorders. Principal among these is Alzheimer's disease. Here we review an already sizeable existing literature on the neuropathological, structural and functional neuroimaging studies of the cerebellum in Alzheimer's disease. We consider these observations in the light of the cognitive deficits that characterize Alzheimer's disease and in so doing we introduce a new perspective on its pathophysiology and manifestations. We propose an integrative hypothesis that there is a cerebellar contribution to the cognitive and neuropsychiatric deficits in Alzheimer's disease. We draw on the dysmetria of thought theory to suggest that this cerebellar component manifests as deficits in modulation of the neurobehavioural deficits. We provide suggestions for future studies to investigate this hypothesis and, ultimately, to establish a comprehensive, causal clinicopathological disease model.
Collapse
Affiliation(s)
- Heidi I L Jacobs
- School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, PO BOX 616, 6200 MD, AQ220 Maastricht, The Netherlands.,Faculty of Psychology and Neuroscience, Department of Cognitive Neuroscience, Maastricht University, PO BOX 616, 6200 MD Maastricht, The Netherlands.,Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - David A Hopkins
- School for Mental Health and Neuroscience, Department of Neuroscience, Maastricht University, PO BOX 616, 6200 MD Maastricht, The Netherlands.,Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Helen C Mayrhofer
- Faculty of Psychology and Neuroscience, Department of Cognitive Neuroscience, Maastricht University, PO BOX 616, 6200 MD Maastricht, The Netherlands
| | - Emiliano Bruner
- Centro Nacional de Investigación sobre la Evolución Humana, Burgos, Spain
| | - Fred W van Leeuwen
- School for Mental Health and Neuroscience, Department of Neuroscience, Maastricht University, PO BOX 616, 6200 MD Maastricht, The Netherlands
| | - Wijnand Raaijmakers
- Faculty of Psychology and Neuroscience, Department of Cognitive Neuroscience, Maastricht University, PO BOX 616, 6200 MD Maastricht, The Netherlands
| | - Jeremy D Schmahmann
- Ataxia Unit, Cognitive Behavioral Neurology Unit, Laboratory for Neuroanatomy and Cerebellar Neurobiology, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
25
|
Ramirez Aguilar L, Acosta-Uribe J, Giraldo MM, Moreno S, Baena A, Alzate D, Cuastumal R, Aguillón D, Madrigal L, Saldarriaga A, Navarro A, Garcia GP, Aguirre-Acevedo DC, Geier EG, Cochran JN, Quiroz YT, Myers RM, Yokoyama JS, Kosik KS, Lopera F. Genetic origin of a large family with a novel PSEN1 mutation (Ile416Thr). Alzheimers Dement 2019; 15:709-719. [PMID: 30745123 DOI: 10.1016/j.jalz.2018.12.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 12/14/2018] [Accepted: 12/17/2018] [Indexed: 11/25/2022]
Abstract
INTRODUCTION A small percentage of Alzheimer's disease (AD) cases are caused by genetic mutations with autosomal dominant inheritance. We report a family with a novel variant in PSEN1. METHODS We performed clinical and genetic evaluation of 93 related individuals from a Colombian admixed population. 31 individuals had whole-genome sequencing. RESULTS Genetic analysis revealed a missense variant in PSEN1 (NM_000021.3: c.1247T>C p.Ile416Thr), which originated on an African haplotype and segregated with AD logarithm of the odds score of 6. Their clinical phenotype is similar to sporadic AD except for earlier age at onset: the mean age at onset for mild cognitive impairment was 47.6 years (standard deviation 5.83) and for dementia 51.6 years (standard deviation 5.03). DISCUSSION Ile416Thr is a novel pathogenic variant that causes AD in the sixth decade of life. The history of the region that included slave importation and admixtures within a confined geographic locale represents a "mini-population bottleneck" and subsequent emergence of a rare dominant mutation.
Collapse
Affiliation(s)
- Laura Ramirez Aguilar
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Juliana Acosta-Uribe
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Antioquia, Colombia; Neuroscience Research Institute, University of California, Santa Barbara, CA, USA; Department of Molecular Cellular and Developmental Biology University of California, Santa Barbara, CA, USA
| | - Margarita M Giraldo
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Sonia Moreno
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Ana Baena
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Diana Alzate
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Rosario Cuastumal
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - David Aguillón
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Lucía Madrigal
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Amanda Saldarriaga
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Alexander Navarro
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Gloria P Garcia
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Daniel C Aguirre-Acevedo
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Ethan G Geier
- Department of Neurology, University of California, San Francisco, CA, USA
| | | | - Yakeel T Quiroz
- Departments of Psychiatry and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard M Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | | | - Kenneth S Kosik
- Neuroscience Research Institute, University of California, Santa Barbara, CA, USA; Department of Molecular Cellular and Developmental Biology University of California, Santa Barbara, CA, USA.
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Antioquia, Colombia
| |
Collapse
|
26
|
Nicolas G, Veltman JA. The role of de novo mutations in adult-onset neurodegenerative disorders. Acta Neuropathol 2019; 137:183-207. [PMID: 30478624 PMCID: PMC6513904 DOI: 10.1007/s00401-018-1939-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/14/2018] [Accepted: 11/14/2018] [Indexed: 12/13/2022]
Abstract
The genetic underpinnings of the most common adult-onset neurodegenerative disorders (AOND) are complex in majority of the cases. In some families, however, the disease can be inherited in a Mendelian fashion as an autosomal-dominant trait. Next to that, patients carrying mutations in the same disease genes have been reported despite a negative family history. Although challenging to demonstrate due to the late onset of the disease in most cases, the occurrence of de novo mutations can explain this sporadic presentation, as demonstrated for severe neurodevelopmental disorders. Exome or genome sequencing of patient-parent trios allows a hypothesis-free study of the role of de novo mutations in AOND and the discovery of novel disease genes. Another hypothesis that may explain a proportion of sporadic AOND cases is the occurrence of a de novo mutation after the fertilization of the oocyte (post-zygotic mutation) or even as a late-somatic mutation, restricted to the brain. Such somatic mutation hypothesis, that can be tested with the use of novel sequencing technologies, is fully compatible with the seeding and spreading mechanisms of the pathological proteins identified in most of these disorders. We review here the current knowledge and future perspectives on de novo mutations in known and novel candidate genes identified in the most common AONDs such as Alzheimer's disease, Parkinson's disease, the frontotemporal lobar degeneration spectrum and Prion disorders. Also, we review the first lessons learned from recent genomic studies of control and diseased brains and the challenges which remain to be addressed.
Collapse
Affiliation(s)
- Gaël Nicolas
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Genetics and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, 22, Boulevard Gambetta, 76000, 76031, Rouen Cedex, France.
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Joris A Veltman
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
27
|
Vega IE, Cabrera LY, Wygant CM, Velez-Ortiz D, Counts SE. Alzheimer's Disease in the Latino Community: Intersection of Genetics and Social Determinants of Health. J Alzheimers Dis 2018; 58:979-992. [PMID: 28527211 DOI: 10.3233/jad-161261] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia among individuals 65 or older. There are more than 5 million diagnosed cases in the US alone and this number is expected to triple by 2050. Therefore, AD has reached epidemic proportions with significant socioeconomic implications. While aging in general is the greatest risk factor for AD, several additional demographic factors that have contributed to the rise in AD in the US are under study. One such factor is associated with the relatively fast growth of the Latino population. Several reports indicate that AD is more prevalent among blacks and Latinos. However, the reason for AD disparity among different ethnic groups is still poorly understood and highly controversial. The Latino population is composed of different groups based on nationality, namely South and Central America, Mexico, and Caribbean Hispanics. This diversity among the Latino population represents an additional challenge since there are distinct characteristics associated with AD and comorbidities. In this review, we aim to bring attention to the intersection between social determinants of health and genetic factors associated with AD within the Latino community. We argue that understanding the interplay between identified social determinants of health, co-morbidities, and genetic factors could lead to community empowerment and inclusiveness in research and healthcare services, contributing to improved diagnosis and treatment of AD patients. Lastly, we propose that inserting a neuroethics perspective could help understand key challenges that influence healthcare disparities and contribute to increased risk of AD among Latinos.
Collapse
Affiliation(s)
- Irving E Vega
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.,Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Laura Y Cabrera
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.,Center for Ethics and Humanities in the Life Sciences, Michigan State University, East Lansing, MI, USA
| | - Cassandra M Wygant
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | | | - Scott E Counts
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.,Department of Family Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.,Hauenstein Neurosciences Center, Mercy Health Saint Mary's Hospital, Grand Rapids, MI, USA
| |
Collapse
|
28
|
Belbin GM, Odgis J, Sorokin EP, Yee MC, Kohli S, Glicksberg BS, Gignoux CR, Wojcik GL, Van Vleck T, Jeff JM, Linderman M, Schurmann C, Ruderfer D, Cai X, Merkelson A, Justice AE, Young KL, Graff M, North KE, Peters U, James R, Hindorff L, Kornreich R, Edelmann L, Gottesman O, Stahl EE, Cho JH, Loos RJ, Bottinger EP, Nadkarni GN, Abul-Husn NS, Kenny EE. Genetic identification of a common collagen disease in puerto ricans via identity-by-descent mapping in a health system. eLife 2017; 6:25060. [PMID: 28895531 PMCID: PMC5595434 DOI: 10.7554/elife.25060] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 08/09/2017] [Indexed: 11/16/2022] Open
Abstract
Achieving confidence in the causality of a disease locus is a complex task that often requires supporting data from both statistical genetics and clinical genomics. Here we describe a combined approach to identify and characterize a genetic disorder that leverages distantly related patients in a health system and population-scale mapping. We utilize genomic data to uncover components of distant pedigrees, in the absence of recorded pedigree information, in the multi-ethnic BioMe biobank in New York City. By linking to medical records, we discover a locus associated with both elevated genetic relatedness and extreme short stature. We link the gene, COL27A1, with a little-known genetic disease, previously thought to be rare and recessive. We demonstrate that disease manifests in both heterozygotes and homozygotes, indicating a common collagen disorder impacting up to 2% of individuals of Puerto Rican ancestry, leading to a better understanding of the continuum of complex and Mendelian disease. Diseases often run in families. These disease are frequently linked to changes in DNA that are passed down through generations. Close family members may share these disease-causing mutations; so may distant relatives who inherited the same mutation from a common ancestor long ago. Geneticists use a method called linkage mapping to trace a disease found in multiple members of a family over generations to genetic changes in a shared ancestor. This allows scientists to pinpoint the exact place in the genome the disease-causing mutation occurred. Using computer algorithms, scientists can apply the same technique to identify mutations that distant relatives inherited from a common ancestor. Belbin et al. used this computational technique to identify a mutation that may cause unusually short stature or bone and joint problems in up to 2% of people of Puerto Rican descent. In the experiments, the genomes of about 32,000 New Yorkers who have volunteered to participate in the BioMe Biobank and their health records were used to search for genetic changes linked to extremely short stature. The search revealed that people who inherited two copies of this mutation from their parents were likely to be extremely short or to have bone and joint problems. People who inherited one copy had an increased likelihood of joint or bone problems. This mutation affects a gene responsible for making a form of protein called collagen that is important for bone growth. The analysis suggests the mutation first arose in a Native American ancestor living in Puerto Rico around the time that European colonization began. The mutation had previously been linked to a disorder called Steel syndrome that was thought to be rare. Belbin et al. showed this condition is actually fairly common in people whose ancestors recently came from Puerto Rico, but may often go undiagnosed by their physicians. The experiments emphasize the importance of including diverse populations in genetic studies, as studies of people of predominantly European descent would likely have missed the link between this disease and mutation.
Collapse
Affiliation(s)
- Gillian Morven Belbin
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, United States.,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, United States.,The Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Jacqueline Odgis
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Elena P Sorokin
- Department of Genetics, Stanford University School of Medicine, Stanford, United States
| | - Muh-Ching Yee
- Department of Plant Biology, Carnegie Institution for Science, Stanford, United States
| | - Sumita Kohli
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Benjamin S Glicksberg
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, United States.,The Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, United States.,Harris Center for Precision Wellness, Icahn School of Medicine at Mt Sinai, New York, United States
| | - Christopher R Gignoux
- Department of Genetics, Stanford University School of Medicine, Stanford, United States
| | - Genevieve L Wojcik
- Department of Genetics, Stanford University School of Medicine, Stanford, United States
| | - Tielman Van Vleck
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Janina M Jeff
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Michael Linderman
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, United States.,The Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Claudia Schurmann
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Douglas Ruderfer
- Broad Institute, Cambridge, United States.,Division of Psychiatric Genomics, Icahn School of Medicine at Mt Sinai, New York, United States.,Center for Statistical Genetics, Icahn School of Medicine at Mt Sinai, New York, United States
| | - Xiaoqiang Cai
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Amanda Merkelson
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Anne E Justice
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Kristin L Young
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Misa Graff
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Kari E North
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, United States.,Department of Epidemiology, University of Washington School of Public Health, Seattle, United States
| | - Regina James
- National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, United States
| | - Lucia Hindorff
- National Human Genome Research Institute, National Institutes of Health, Bethesda, United States
| | - Ruth Kornreich
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Lisa Edelmann
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Omri Gottesman
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Eli Ea Stahl
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, United States.,The Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, United States.,Harris Center for Precision Wellness, Icahn School of Medicine at Mt Sinai, New York, United States.,Broad Institute, Cambridge, United States
| | - Judy H Cho
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, United States.,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, United States.,Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Ruth Jf Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, United States.,The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Erwin P Bottinger
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Girish N Nadkarni
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Noura S Abul-Husn
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, United States.,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, United States.,The Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Eimear E Kenny
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, United States.,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, United States.,The Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, United States.,Center for Statistical Genetics, Icahn School of Medicine at Mt Sinai, New York, United States
| |
Collapse
|
29
|
Van Horn JD, Irimia A, Torgerson CM, Bhattrai A, Jacokes Z, Vespa PM. Mild cognitive impairment and structural brain abnormalities in a sexagenarian with a history of childhood traumatic brain injury. J Neurosci Res 2017; 96:652-660. [PMID: 28543689 DOI: 10.1002/jnr.24084] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 12/30/2022]
Abstract
In this report, we present a case study involving an older, female patient with a history of pediatric traumatic brain injury (TBI). Magnetic resonance imaging and diffusion tensor imaging volumes were acquired from the volunteer in question, her brain volumetrics and morphometrics were extracted, and these were then systematically compared against corresponding metrics obtained from a large sample of older healthy control (HC) subjects as well as from subjects in various stages of mild cognitive impairment (MCI) and Alzheimer disease (AD). Our analyses find the patient's brain morphometry and connectivity most similar to those of patients classified as having early-onset MCI, in contrast to HC, late MCI, and AD samples. Our examination will be of particular interest to those interested in assessing the clinical course in older patients having suffered TBI earlier in life, in contradistinction to those who experience incidents of head injury during aging.
Collapse
Affiliation(s)
- John Darrell Van Horn
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, California
| | - Andrei Irimia
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, California
| | - Carinna M Torgerson
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, California
| | - Avnish Bhattrai
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, California
| | - Zachary Jacokes
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, California
| | - Paul M Vespa
- Department of Neurosurgery, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
30
|
|
31
|
A Mutation in DAOA Modifies the Age of Onset in PSEN1 E280A Alzheimer's Disease. Neural Plast 2016; 2016:9760314. [PMID: 26949549 PMCID: PMC4753688 DOI: 10.1155/2016/9760314] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 09/30/2015] [Accepted: 10/21/2015] [Indexed: 11/17/2022] Open
Abstract
We previously reported age of onset (AOO) modifier genes in the world's largest pedigree segregating early-onset Alzheimer's disease (AD), caused by the p.Glu280Ala (E280A) mutation in the PSEN1 gene. Here we report the results of a targeted analysis of functional exonic variants in those AOO modifier genes in sixty individuals with PSEN1 E280A AD who were whole-exome genotyped for ~250,000 variants. Standard quality control, filtering, and annotation for functional variants were applied, and common functional variants located in those previously reported as AOO modifier loci were selected. Multiloci linear mixed-effects models were used to test the association between these variants and AOO. An exonic missense mutation in the G72 (DAOA) gene (rs2391191, P = 1.94 × 10−4, PFDR = 9.34 × 10−3) was found to modify AOO in PSEN1 E280A AD. Nominal associations of missense mutations in the CLUAP1 (rs9790, P = 7.63 × 10−3, PFDR = 0.1832) and EXOC2 (rs17136239, P = 0.0325, PFDR = 0.391) genes were also found. Previous studies have linked polymorphisms in the DAOA gene with the occurrence of neuropsychiatric symptoms such as depression, apathy, aggression, delusions, hallucinations, and psychosis in AD. Our findings strongly suggest that this new conspicuous functional AOO modifier within the G72 (DAOA) gene could be pivotal for understanding the genetic basis of AD.
Collapse
|
32
|
Zeevi DA, Altarescu G, Weinberg-Shukron A, Zahdeh F, Dinur T, Chicco G, Herskovitz Y, Renbaum P, Elstein D, Levy-Lahad E, Rolfs A, Zimran A. Proof-of-principle rapid noninvasive prenatal diagnosis of autosomal recessive founder mutations. J Clin Invest 2015; 125:3757-65. [PMID: 26426075 DOI: 10.1172/jci79322] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 07/23/2015] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Noninvasive prenatal testing can be used to accurately detect chromosomal aneuploidies in circulating fetal DNA; however, the necessity of parental haplotype construction is a primary drawback to noninvasive prenatal diagnosis (NIPD) of monogenic disease. Family-specific haplotype assembly is essential for accurate diagnosis of minuscule amounts of circulating cell-free fetal DNA; however, current haplotyping techniques are too time-consuming and laborious to be carried out within the limited time constraints of prenatal testing, hampering practical application of NIPD in the clinic. Here, we have addressed this pitfall and devised a universal strategy for rapid NIPD of a prevalent mutation in the Ashkenazi Jewish (AJ) population. METHODS Pregnant AJ couples, carrying mutation(s) in GBA, which encodes acid β-glucosidase, were recruited at the SZMC Gaucher Clinic. Targeted next-generation sequencing of GBA-flanking SNPs was performed on peripheral blood samples from each couple, relevant mutation carrier family members, and unrelated individuals who are homozygotes for an AJ founder mutation. Allele-specific haplotypes were constructed based on linkage, and a consensus Gaucher disease-associated founder mutation-flanking haplotype was fine mapped. Together, these haplotypes were used for NIPD. All test results were validated by conventional prenatal or postnatal diagnostic methods. RESULTS Ten parental alleles in eight unrelated fetuses were diagnosed successfully based on the noninvasive method developed in this study. The consensus mutation-flanking haplotype aided diagnosis for 6 of 9 founder mutation alleles. CONCLUSIONS The founder NIPD method developed and described here is rapid, economical, and readily adaptable for prenatal testing of prevalent autosomal recessive disease-causing mutations in an assortment of worldwide populations. FUNDING SZMC, Protalix Biotherapeutics Inc., and Centogene AG.
Collapse
|
33
|
Schindler SE, Fagan AM. Autosomal Dominant Alzheimer Disease: A Unique Resource to Study CSF Biomarker Changes in Preclinical AD. Front Neurol 2015; 6:142. [PMID: 26175713 PMCID: PMC4483518 DOI: 10.3389/fneur.2015.00142] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 06/12/2015] [Indexed: 12/27/2022] Open
Abstract
Our understanding of the pathogenesis of Alzheimer disease (AD) has been greatly influenced by investigation of rare families with autosomal dominant mutations that cause early onset AD. Mutations in the genes coding for amyloid precursor protein (APP), presenilin 1 (PSEN-1), and presenilin 2 (PSEN-2) cause over-production of the amyloid-β peptide (Aβ) leading to early deposition of Aβ in the brain, which in turn is hypothesized to initiate a cascade of processes, resulting in neuronal death, cognitive decline, and eventual dementia. Studies of cerebrospinal fluid (CSF) from individuals with the common form of AD, late-onset AD (LOAD), have revealed that low CSF Aβ42 and high CSF tau are associated with AD brain pathology. Herein, we review the literature on CSF biomarkers in autosomal dominant AD (ADAD), which has contributed to a detailed road map of AD pathogenesis, especially during the preclinical period, prior to the appearance of any cognitive symptoms. Current drug trials are also taking advantage of the unique characteristics of ADAD and utilizing CSF biomarkers to accelerate development of effective therapies for AD.
Collapse
Affiliation(s)
- Suzanne Elizabeth Schindler
- Department of Neurology, Knight Alzheimer's Disease Research Center, Hope Center for Neurological Disorders, Washington University School of Medicine , St. Louis, MO , USA
| | - Anne M Fagan
- Department of Neurology, Knight Alzheimer's Disease Research Center, Hope Center for Neurological Disorders, Washington University School of Medicine , St. Louis, MO , USA
| |
Collapse
|
34
|
Abstract
Alzheimer's disease (AD), the most common form of dementia in western societies, is a pathologically and clinically heterogeneous disease with a strong genetic component. The recent advances in high-throughput genome technologies allowing for the rapid analysis of millions of polymorphisms in thousands of subjects has significantly advanced our understanding of the genomic underpinnings of AD susceptibility. During the last 5 years, genome-wide association and whole-exome- and whole-genome sequencing studies have mapped more than 20 disease-associated loci, providing insights into the molecular pathways involved in AD pathogenesis and hinting at potential novel therapeutic targets. This review article summarizes the challenges and opportunities of when using genomic information for the diagnosis and prognosis of AD.
Collapse
Affiliation(s)
- Christiane Reitz
- Sergievsly Center/Taub Institute/Dept. of Neurology, Columbia University, 630 W 168th Street, Rm 19-308, New York, NY 10032, phone: (212) 305-0865, fax: (212) 305-2391
| |
Collapse
|
35
|
Jimenez-Del-Rio M, Velez-Pardo C. Alzheimer’s Disease, Drosophila melanogaster and Polyphenols. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 863:21-53. [DOI: 10.1007/978-3-319-18365-7_2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
36
|
Kosik KS, Muñoz C, Lopez L, Arcila ML, García G, Madrigal L, Moreno S, Ríos Romenets S, Lopez H, Gutierrez M, Langbaum JB, Cho W, Suliman S, Tariot PN, Ho C, Reiman EM, Lopera F. Homozygosity of the autosomal dominant Alzheimer disease presenilin 1 E280A mutation. Neurology 2014; 84:206-8. [PMID: 25471389 DOI: 10.1212/wnl.0000000000001130] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Kenneth S Kosik
- From the Neuroscience Research Institute (K.S.K., M.L.A.), University of California, Santa Barbara; Grupo de Neurociencias de Antioquia (C.M., L.L., G.G., L.M., S.M., S.R.R., H.L., M.G., F.L.), Universidad de Antioquia, Medellín, Colombia; Banner Alzheimer's Institute (J.B.L., P.N.T., E.M.R.), Phoenix, AZ; and Genentech (W.C., S.S., C.H.), South San Francisco, CA.
| | - Claudia Muñoz
- From the Neuroscience Research Institute (K.S.K., M.L.A.), University of California, Santa Barbara; Grupo de Neurociencias de Antioquia (C.M., L.L., G.G., L.M., S.M., S.R.R., H.L., M.G., F.L.), Universidad de Antioquia, Medellín, Colombia; Banner Alzheimer's Institute (J.B.L., P.N.T., E.M.R.), Phoenix, AZ; and Genentech (W.C., S.S., C.H.), South San Francisco, CA
| | - Liliana Lopez
- From the Neuroscience Research Institute (K.S.K., M.L.A.), University of California, Santa Barbara; Grupo de Neurociencias de Antioquia (C.M., L.L., G.G., L.M., S.M., S.R.R., H.L., M.G., F.L.), Universidad de Antioquia, Medellín, Colombia; Banner Alzheimer's Institute (J.B.L., P.N.T., E.M.R.), Phoenix, AZ; and Genentech (W.C., S.S., C.H.), South San Francisco, CA
| | - Mary Luz Arcila
- From the Neuroscience Research Institute (K.S.K., M.L.A.), University of California, Santa Barbara; Grupo de Neurociencias de Antioquia (C.M., L.L., G.G., L.M., S.M., S.R.R., H.L., M.G., F.L.), Universidad de Antioquia, Medellín, Colombia; Banner Alzheimer's Institute (J.B.L., P.N.T., E.M.R.), Phoenix, AZ; and Genentech (W.C., S.S., C.H.), South San Francisco, CA
| | - Gloria García
- From the Neuroscience Research Institute (K.S.K., M.L.A.), University of California, Santa Barbara; Grupo de Neurociencias de Antioquia (C.M., L.L., G.G., L.M., S.M., S.R.R., H.L., M.G., F.L.), Universidad de Antioquia, Medellín, Colombia; Banner Alzheimer's Institute (J.B.L., P.N.T., E.M.R.), Phoenix, AZ; and Genentech (W.C., S.S., C.H.), South San Francisco, CA
| | - Lucía Madrigal
- From the Neuroscience Research Institute (K.S.K., M.L.A.), University of California, Santa Barbara; Grupo de Neurociencias de Antioquia (C.M., L.L., G.G., L.M., S.M., S.R.R., H.L., M.G., F.L.), Universidad de Antioquia, Medellín, Colombia; Banner Alzheimer's Institute (J.B.L., P.N.T., E.M.R.), Phoenix, AZ; and Genentech (W.C., S.S., C.H.), South San Francisco, CA
| | - Sonia Moreno
- From the Neuroscience Research Institute (K.S.K., M.L.A.), University of California, Santa Barbara; Grupo de Neurociencias de Antioquia (C.M., L.L., G.G., L.M., S.M., S.R.R., H.L., M.G., F.L.), Universidad de Antioquia, Medellín, Colombia; Banner Alzheimer's Institute (J.B.L., P.N.T., E.M.R.), Phoenix, AZ; and Genentech (W.C., S.S., C.H.), South San Francisco, CA
| | - Silvia Ríos Romenets
- From the Neuroscience Research Institute (K.S.K., M.L.A.), University of California, Santa Barbara; Grupo de Neurociencias de Antioquia (C.M., L.L., G.G., L.M., S.M., S.R.R., H.L., M.G., F.L.), Universidad de Antioquia, Medellín, Colombia; Banner Alzheimer's Institute (J.B.L., P.N.T., E.M.R.), Phoenix, AZ; and Genentech (W.C., S.S., C.H.), South San Francisco, CA
| | - Hugo Lopez
- From the Neuroscience Research Institute (K.S.K., M.L.A.), University of California, Santa Barbara; Grupo de Neurociencias de Antioquia (C.M., L.L., G.G., L.M., S.M., S.R.R., H.L., M.G., F.L.), Universidad de Antioquia, Medellín, Colombia; Banner Alzheimer's Institute (J.B.L., P.N.T., E.M.R.), Phoenix, AZ; and Genentech (W.C., S.S., C.H.), South San Francisco, CA
| | - Madelyn Gutierrez
- From the Neuroscience Research Institute (K.S.K., M.L.A.), University of California, Santa Barbara; Grupo de Neurociencias de Antioquia (C.M., L.L., G.G., L.M., S.M., S.R.R., H.L., M.G., F.L.), Universidad de Antioquia, Medellín, Colombia; Banner Alzheimer's Institute (J.B.L., P.N.T., E.M.R.), Phoenix, AZ; and Genentech (W.C., S.S., C.H.), South San Francisco, CA
| | - Jessica B Langbaum
- From the Neuroscience Research Institute (K.S.K., M.L.A.), University of California, Santa Barbara; Grupo de Neurociencias de Antioquia (C.M., L.L., G.G., L.M., S.M., S.R.R., H.L., M.G., F.L.), Universidad de Antioquia, Medellín, Colombia; Banner Alzheimer's Institute (J.B.L., P.N.T., E.M.R.), Phoenix, AZ; and Genentech (W.C., S.S., C.H.), South San Francisco, CA
| | - William Cho
- From the Neuroscience Research Institute (K.S.K., M.L.A.), University of California, Santa Barbara; Grupo de Neurociencias de Antioquia (C.M., L.L., G.G., L.M., S.M., S.R.R., H.L., M.G., F.L.), Universidad de Antioquia, Medellín, Colombia; Banner Alzheimer's Institute (J.B.L., P.N.T., E.M.R.), Phoenix, AZ; and Genentech (W.C., S.S., C.H.), South San Francisco, CA
| | - Shehnaaz Suliman
- From the Neuroscience Research Institute (K.S.K., M.L.A.), University of California, Santa Barbara; Grupo de Neurociencias de Antioquia (C.M., L.L., G.G., L.M., S.M., S.R.R., H.L., M.G., F.L.), Universidad de Antioquia, Medellín, Colombia; Banner Alzheimer's Institute (J.B.L., P.N.T., E.M.R.), Phoenix, AZ; and Genentech (W.C., S.S., C.H.), South San Francisco, CA
| | - Pierre N Tariot
- From the Neuroscience Research Institute (K.S.K., M.L.A.), University of California, Santa Barbara; Grupo de Neurociencias de Antioquia (C.M., L.L., G.G., L.M., S.M., S.R.R., H.L., M.G., F.L.), Universidad de Antioquia, Medellín, Colombia; Banner Alzheimer's Institute (J.B.L., P.N.T., E.M.R.), Phoenix, AZ; and Genentech (W.C., S.S., C.H.), South San Francisco, CA
| | - Carole Ho
- From the Neuroscience Research Institute (K.S.K., M.L.A.), University of California, Santa Barbara; Grupo de Neurociencias de Antioquia (C.M., L.L., G.G., L.M., S.M., S.R.R., H.L., M.G., F.L.), Universidad de Antioquia, Medellín, Colombia; Banner Alzheimer's Institute (J.B.L., P.N.T., E.M.R.), Phoenix, AZ; and Genentech (W.C., S.S., C.H.), South San Francisco, CA
| | - Eric M Reiman
- From the Neuroscience Research Institute (K.S.K., M.L.A.), University of California, Santa Barbara; Grupo de Neurociencias de Antioquia (C.M., L.L., G.G., L.M., S.M., S.R.R., H.L., M.G., F.L.), Universidad de Antioquia, Medellín, Colombia; Banner Alzheimer's Institute (J.B.L., P.N.T., E.M.R.), Phoenix, AZ; and Genentech (W.C., S.S., C.H.), South San Francisco, CA
| | - Francisco Lopera
- From the Neuroscience Research Institute (K.S.K., M.L.A.), University of California, Santa Barbara; Grupo de Neurociencias de Antioquia (C.M., L.L., G.G., L.M., S.M., S.R.R., H.L., M.G., F.L.), Universidad de Antioquia, Medellín, Colombia; Banner Alzheimer's Institute (J.B.L., P.N.T., E.M.R.), Phoenix, AZ; and Genentech (W.C., S.S., C.H.), South San Francisco, CA
| |
Collapse
|