1
|
Campanelli L, Galeano P, Prestia FA, Cuesta C, Dalmasso MC, Flores-López M, Gona C, Irureta N, Kairiyama C, Lisso J, López-Gambero AJ, Mintz I, Medel N, Campuzano KS, Muchnik C, Novack GV, Olivar N, Quiroga I, Requena-Ocaña N, Reyes-Bueno JA, Serrano-Castro P, Sevillano Z, Solis P, Suárez J, Villella I, Wukitsevits N, Castaño EM, Taragano F, Kochen S, Politis DG, Brusco LI, Rodríguez de Fonseca F, Morelli L. Blood levels of cytokines highlight the role of inflammation in Alzheimer's disease. Heliyon 2025; 11:e41725. [PMID: 39872450 PMCID: PMC11770505 DOI: 10.1016/j.heliyon.2025.e41725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/27/2024] [Accepted: 01/03/2025] [Indexed: 01/30/2025] Open
Abstract
Inflammation and angiogenesis have been defined as potential mechanisms associated with clinical progression from a cognitively normal state to Alzheimer's disease (AD). In this observational case-control study, we aimed to determine plasma levels of cytokines as indicators of inflammation involved in cognitive decline. We measured 30 plasma proteins in 49 controls (CTL), 36 individuals with mild cognitive impairment (MCI) and 52 patients diagnosed with probable AD. After applying strict filters for quantification limits, only 13 analytes were included in the analysis. Kruskal-Wallis tests showed significant differences between diagnostic groups for nine cytokines (IL-16, IL-7, VEGF, IL-8, eotaxin, MCP-1, MCP-4, MDC and TARC). Non-parametric MANCOVA showed that sex and diagnosis affected cytokine levels in the blood. To determine the sensitivity and specificity of the markers, we performed receiver operating characteristic (ROC) curve analysis. Only those analytes that showed an area under the curve (AUC) ≥ 0.70 were included in the multivariate logistic regression models to better understand the contribution of cytokines to clinical status. Three models: 1) CTL vs. AD; 2) CTL vs. MCI, and 3) MCI vs. AD were developed, with sex and age as covariates. In each model, two cytokines remained significantly different (model 1: IL-16 and MDC; model 2: eotaxin and MDC and model 3: IL-7 and VEGF). Taken together, this report identifies a set of plasma markers of inflammation and strengthens the role of glial biology in different clinical stages of AD.
Collapse
Affiliation(s)
- Lorenzo Campanelli
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir, IIBBA-CONICET, Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires, Argentina
| | - Pablo Galeano
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir, IIBBA-CONICET, Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires, Argentina
| | - Federico A. Prestia
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir, IIBBA-CONICET, Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires, Argentina
| | - Carolina Cuesta
- Hospital Interzonal General de Agudos (HIGA) Eva Perón, Av. 101 Dr. Ricardo Balbín 3200, Provincia de Buenos Aires, Buenos Aires, B1650NBN, Argentina
| | - Maria C. Dalmasso
- Studies in Neuroscience and Complex Systems Unit (CONICET-HEC-UNAJ), Av. Calchaquí 5402, Florencio Varela, Argentina
| | - María Flores-López
- Grupo de Neuropsicofarmacología, IBIMA-Plataforma BIONAND, Hospital Regional Universitario de Málaga, Puerto de la Torre, 29010, Málaga, Spain
- Unidad de Gestión Clínica de Salud Mental, IBIMA-Plataforma BIONAND, Hospital Regional Universitario de Málaga, Málaga, Spain, Puerto de la Torre, 29010, Málaga, Spain
| | - Cristian Gona
- Asistencia Medica Integral, Hospital El Cruce, Av. Calchaquí 5401, Provincia de Buenos Aires, Florencio Varela, Argentina
| | - Nicolás Irureta
- Studies in Neuroscience and Complex Systems Unit (CONICET-HEC-UNAJ), Av. Calchaquí 5402, Florencio Varela, Argentina
- Asistencia Medica Integral, Hospital El Cruce, Av. Calchaquí 5401, Provincia de Buenos Aires, Florencio Varela, Argentina
| | - Claudia Kairiyama
- Hospital Interzonal General de Agudos (HIGA) Eva Perón, Av. 101 Dr. Ricardo Balbín 3200, Provincia de Buenos Aires, Buenos Aires, B1650NBN, Argentina
| | - Julieta Lisso
- Studies in Neuroscience and Complex Systems Unit (CONICET-HEC-UNAJ), Av. Calchaquí 5402, Florencio Varela, Argentina
- Asistencia Medica Integral, Hospital El Cruce, Av. Calchaquí 5401, Provincia de Buenos Aires, Florencio Varela, Argentina
| | - Antonio Jesús López-Gambero
- Grupo de Neuropsicofarmacología, IBIMA-Plataforma BIONAND, Hospital Regional Universitario de Málaga, Puerto de la Torre, 29010, Málaga, Spain
- Unidad de Gestión Clínica de Salud Mental, IBIMA-Plataforma BIONAND, Hospital Regional Universitario de Málaga, Málaga, Spain, Puerto de la Torre, 29010, Málaga, Spain
| | - Ines Mintz
- Studies in Neuroscience and Complex Systems Unit (CONICET-HEC-UNAJ), Av. Calchaquí 5402, Florencio Varela, Argentina
- Asistencia Medica Integral, Hospital El Cruce, Av. Calchaquí 5401, Provincia de Buenos Aires, Florencio Varela, Argentina
| | - Nancy Medel
- Studies in Neuroscience and Complex Systems Unit (CONICET-HEC-UNAJ), Av. Calchaquí 5402, Florencio Varela, Argentina
- Asistencia Medica Integral, Hospital El Cruce, Av. Calchaquí 5401, Provincia de Buenos Aires, Florencio Varela, Argentina
| | - Karen S. Campuzano
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir, IIBBA-CONICET, Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires, Argentina
| | - Carolina Muchnik
- Center of Neuropsychiatry and Behavior Neurology, School of Medicine, University of Buenos Aires, Paraguay 2155, Ciudad Autónoma de Buenos Aires, Argentina
| | - Gisela V. Novack
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir, IIBBA-CONICET, Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires, Argentina
| | - Natividad Olivar
- Center of Neuropsychiatry and Behavior Neurology, School of Medicine, University of Buenos Aires, Paraguay 2155, Ciudad Autónoma de Buenos Aires, Argentina
| | - Ivana Quiroga
- Asistencia Medica Integral, Hospital El Cruce, Av. Calchaquí 5401, Provincia de Buenos Aires, Florencio Varela, Argentina
| | - Nerea Requena-Ocaña
- Grupo de Neuropsicofarmacología, IBIMA-Plataforma BIONAND, Hospital Regional Universitario de Málaga, Puerto de la Torre, 29010, Málaga, Spain
- Unidad de Gestión Clínica de Salud Mental, IBIMA-Plataforma BIONAND, Hospital Regional Universitario de Málaga, Málaga, Spain, Puerto de la Torre, 29010, Málaga, Spain
| | - Jose Antonio Reyes-Bueno
- Unidad de Neurología, IBIMA-Plataforma BIONAND, Hospital Regional Universitario de Málaga, Malaga, Puerto de la Torre, 29010, Málaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology [NEURO-RECA], Av. Carlos Haya, 88, Pabellon B, 4 planta, 29010, Málaga, Spain
| | - Pedro Serrano-Castro
- Unidad de Neurología, IBIMA-Plataforma BIONAND, Hospital Regional Universitario de Málaga, Malaga, Puerto de la Torre, 29010, Málaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology [NEURO-RECA], Av. Carlos Haya, 88, Pabellon B, 4 planta, 29010, Málaga, Spain
| | - Zulma Sevillano
- Studies in Neuroscience and Complex Systems Unit (CONICET-HEC-UNAJ), Av. Calchaquí 5402, Florencio Varela, Argentina
- Asistencia Medica Integral, Hospital El Cruce, Av. Calchaquí 5401, Provincia de Buenos Aires, Florencio Varela, Argentina
| | - Patricia Solis
- Studies in Neuroscience and Complex Systems Unit (CONICET-HEC-UNAJ), Av. Calchaquí 5402, Florencio Varela, Argentina
- Asistencia Medica Integral, Hospital El Cruce, Av. Calchaquí 5401, Provincia de Buenos Aires, Florencio Varela, Argentina
| | - Juan Suárez
- Grupo de Neuropsicofarmacología, IBIMA-Plataforma BIONAND, Hospital Regional Universitario de Málaga, Puerto de la Torre, 29010, Málaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology [NEURO-RECA], Av. Carlos Haya, 88, Pabellon B, 4 planta, 29010, Málaga, Spain
- Departamento of Anatomía Humana, Medicina Legal e Historia de la Ciencia, Facultad de Medicina, Universidad de Málaga, Av. Cervantes, 2, 2907, Málaga, Spain
| | - Ivana Villella
- Studies in Neuroscience and Complex Systems Unit (CONICET-HEC-UNAJ), Av. Calchaquí 5402, Florencio Varela, Argentina
- Asistencia Medica Integral, Hospital El Cruce, Av. Calchaquí 5401, Provincia de Buenos Aires, Florencio Varela, Argentina
| | - Nancy Wukitsevits
- Asistencia Medica Integral, Hospital El Cruce, Av. Calchaquí 5401, Provincia de Buenos Aires, Florencio Varela, Argentina
| | - Eduardo M. Castaño
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir, IIBBA-CONICET, Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires, Argentina
| | - Fernando Taragano
- Neuropsychiatric Clinic Nuestra Señora de Las Nieves, Av. Álvarez Thomas 268, C1427CCP, Ciudad Autónoma de Buenos Aires, Argentina
| | - Silvia Kochen
- Studies in Neuroscience and Complex Systems Unit (CONICET-HEC-UNAJ), Av. Calchaquí 5402, Florencio Varela, Argentina
| | - Daniel G. Politis
- Hospital Interzonal General de Agudos (HIGA) Eva Perón, Av. 101 Dr. Ricardo Balbín 3200, Provincia de Buenos Aires, Buenos Aires, B1650NBN, Argentina
| | - Luis I. Brusco
- Center of Neuropsychiatry and Behavior Neurology, School of Medicine, University of Buenos Aires, Paraguay 2155, Ciudad Autónoma de Buenos Aires, Argentina
| | - Fernando Rodríguez de Fonseca
- Grupo de Neuropsicofarmacología, IBIMA-Plataforma BIONAND, Hospital Regional Universitario de Málaga, Puerto de la Torre, 29010, Málaga, Spain
- Unidad de Gestión Clínica de Salud Mental, IBIMA-Plataforma BIONAND, Hospital Regional Universitario de Málaga, Málaga, Spain, Puerto de la Torre, 29010, Málaga, Spain
- Unidad de Neurología, IBIMA-Plataforma BIONAND, Hospital Regional Universitario de Málaga, Malaga, Puerto de la Torre, 29010, Málaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology [NEURO-RECA], Av. Carlos Haya, 88, Pabellon B, 4 planta, 29010, Málaga, Spain
| | - Laura Morelli
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir, IIBBA-CONICET, Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
2
|
Evans AK, Park HH, Woods CE, Lam RK, Rijsketic DR, Xu C, Chu EK, Ciari P, Blumenfeld S, Vidano LM, Saw NL, Heifets BD, Shamloo M. Impact of noradrenergic inhibition on neuroinflammation and pathophysiology in mouse models of Alzheimer's disease. J Neuroinflammation 2024; 21:322. [PMID: 39696597 PMCID: PMC11657531 DOI: 10.1186/s12974-024-03306-1] [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: 10/24/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024] Open
Abstract
Norepinephrine (NE) modulates cognitive function, arousal, attention, and responses to novelty and stress, and it also regulates neuroinflammation. We previously demonstrated behavioral and immunomodulatory effects of beta-adrenergic pharmacology in mouse models of Alzheimer's disease (AD). The current studies were designed to block noradrenergic signaling in 5XFAD mice through (1) chemogenetic inhibition of the locus coeruleus (LC), (2) pharmacologic blocking of β-adrenergic receptors, and (3) conditional deletion of β1- or β2-adrenergic receptors (adrb1 or adrb2) in microglia.First, brain-wide AD pathology was mapped in 3D by imaging immunolabeled, cleared 5XFAD brains to assess the overlap between amyloid beta (Aβ) pathology, reactive microglia, and the loss of tyrosine hydroxylase (TH) expression in the catecholaminergic system. To examine the effects of inhibiting the LC NE system in the 5XFAD model, inhibitory (Gi) DREADD receptors were expressed specifically in LC NE neurons. LC NE neurons were chronically inhibited through the subcutaneous pump administration of the DREADD agonist clozapine-N-oxide (CNO). Plasma and brains were collected for assessment of neuroinflammation and pathology. A separate cohort of 5XFAD mice was chronically dosed with the beta-adrenergic antagonist propranolol or vehicle and evaluated for behavior, as well as post-mortem neuroinflammation and pathology. Finally, we used 5XFAD mice with conditional deletion of either adrb1 or adrb2 in microglia to assess neuroinflammation and pathology mediated by β-adrenergic signaling.Using iDISCO+, light sheet fluorescence microscopy, and novel analyses, we detected widespread microgliosis and Aβ pathology, along with modest TH downregulation in fibers across multiple brain regions, in contrast to the spatially limited TH downregulation observed in neurons. Both chemogenetic inhibition of LC adrenergic signaling and pharmacological inhibition of beta-adrenergic receptors potentiated neuroinflammation without altering Aβ pathology. Conditional deletion of adrb1 in microglia did not affect neuroinflammation. Conditional deletion of adrb2 in microglia attenuated inflammation and pathology in females but had no effect in males. Overall, these data support previous observations demonstrating the immunomodulatory effects of beta-adrenergic signaling in the pathophysiology of brain disorders and suggest that adrenergic receptors on cell types other than microglia, such as astrocytes, may mediate some of the disease-modifying effects of β-adrenergic agonists in the brain.
Collapse
Affiliation(s)
- Andrew K Evans
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Heui Hye Park
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Claire E Woods
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Rachel K Lam
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Daniel Ryskamp Rijsketic
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Christine Xu
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Emily K Chu
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Peter Ciari
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Sarah Blumenfeld
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Laura M Vidano
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Nay Lui Saw
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Boris D Heifets
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305, United States of America
| | - Mehrdad Shamloo
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America.
| |
Collapse
|
3
|
Evans AK, Park HH, Woods CE, Lam RK, Rijsketic DR, Xu C, Chu E, Ciari P, Blumenfeld S, Vidano LM, Saw NL, Heifets BD, Shamloo M. Impact of noradrenergic inhibition on neuroinflammation and pathophysiology in mouse models of Alzheimer's disease. RESEARCH SQUARE 2024:rs.3.rs-5328229. [PMID: 39574895 PMCID: PMC11581111 DOI: 10.21203/rs.3.rs-5328229/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2024]
Abstract
Norepinephrine (NE) modulates cognitive function, arousal, attention, and responses to novelty and stress, and also regulates neuroinflammation. We previously demonstrated behavioral and immunomodulatory effects of beta-adrenergic pharmacology in mouse models of Alzheimer's disease (AD). The current studies were designed to block noradrenergic signaling in 5XFAD mice through 1) chemogenetic inhibition of the locus coeruleus (LC), 2)pharmacologic blocking of β-adrenergic receptors, and 3) conditional deletion of β1- or β2-adrenergic receptors (adrb1 or adrb2) in microglia. First, brain-wide AD pathology was mapped in 3D by imaging immunolabeled, cleared 5XFAD brains to assess the overlap between Aβ pathology, reactive microglia, and the loss of tyrosine hydroxylase (TH) expression in the catecholaminergic system. To examine the effects of inhibiting the LC NE system in the 5XFAD model, inhibitory (Gi) DREADD receptors were expressed specifically in LC NE neurons. LC NE neurons were chronically inhibited through the subcutaneous pump administration of the DREADD agonist clozapine-N-oxide (CNO). Plasma and brains were collected for assessment of neuroinflammation and pathology. A separate cohort of 5XFAD mice was chronically dosed with the beta-adrenergic antagonist propranolol or vehicle and evaluated for behavior, as well as post-mortem neuroinflammation and pathology. Finally, we used 5XFAD mice with conditional deletion of either adrb1 or adrb2 in microglia to assess neuroinflammation and pathology mediated by β-adrenergic signaling. Using iDISCO, light sheet fluorescence microscopy, and novel analyses, we detected widespread microgliosis and amyloid pathology, along with modest TH downregulation in fibers across multiple brain regions, in contrast to the spatially limited TH downregulation observed in neurons. Both chemogenetic inhibition of LC adrenergic signaling and pharmacological inhibition of beta-adrenergic receptors potentiated neuroinflammation without altering amyloid beta pathology. Conditional deletion of adrb1 in microglia did not affect neuroinflammation. Conditional deletion of adrb2 in microglia attenuated inflammation and pathology in females but had no effect in males. Overall, these data support previous observations demonstrating the immunomodulatory effects of beta-adrenergic signaling in the pathophysiology of brain disorders and suggest that adrenergic receptors on cell types other than microglia, such as astrocytes, may predominantly mediate the disease-modifying effects of β-adrenergic agonists in the brain.
Collapse
|
4
|
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
|
5
|
Sillau SH, Coughlan C, Ahmed MM, Nair K, Araya P, Galbraith MD, Bettcher BM, Espinosa JM, Chial HJ, Epperson N, Boyd TD, Potter H. Neuron loss in the brain starts in childhood, increases exponentially with age and is halted by GM-CSF treatment in Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.14.24310223. [PMID: 39072024 PMCID: PMC11275665 DOI: 10.1101/2024.07.14.24310223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Aging increases the risk of neurodegeneration, cognitive decline, and Alzheimer's disease (AD). Currently no means exist to measure neuronal cell death during life or to prevent it. Here we show that cross-sectional measures of human plasma proteins released from dying/damaged neurons (ubiquitin C-terminal hydrolase-L1/UCH-L1 and neurofilament light/NfL) become exponentially higher from age 2-85; UCH-L1 rises faster in females. Glial fibrillary acidic protein (GFAP) concentrations, indicating astrogliosis/inflammation, increase exponentially after age 40. Treatment with human granulocyte-macrophage colony-stimulating factor (GM-CSF/sargramostim) halted neuronal cell death, as evidenced by reduced plasma UCH-L1 concentrations, in AD participants to levels equivalent to those of five-year-old healthy controls. The ability of GM-CSF treatment to reduce neuronal apoptosis was confirmed in a rat model of AD. These findings suggest that the exponential increase in neurodegeneration with age, accelerated by neuroinflammation, may underlie the contribution of aging to cognitive decline and AD and can be halted by GM-CSF/sargramostim treatment.
Collapse
|
6
|
Lee JY, Lim MCX, Koh RY, Tsen MT, Chye SM. Blood-based therapies to combat neurodegenerative diseases. Metab Brain Dis 2024; 39:985-1004. [PMID: 38842660 DOI: 10.1007/s11011-024-01368-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 05/31/2024] [Indexed: 06/07/2024]
Abstract
Neurodegeneration, known as the progressive loss of neurons in terms of their structure and function, is the principal pathophysiological change found in the majority of brain-related disorders. Ageing has been considered the most well-established risk factor in most common neurodegenerative diseases, such as Parkinson's disease (PD) and Alzheimer's disease (AD). There is currently no effective treatment or cure for these diseases; the approved therapeutic options to date are only for palliative care. Ageing and neurodegenerative diseases are closely intertwined; reversing the aspects of brain ageing could theoretically mitigate age-related neurodegeneration. Ever since the regenerative properties of young blood on aged tissues came to light, substantial efforts have been focused on identifying and characterizing the circulating factors in the young and old systemic milieu that may attenuate or accentuate brain ageing and neurodegeneration. Later studies discovered the superiority of old plasma dilution in tissue rejuvenation, which is achieved through a molecular reset of the systemic proteome. These findings supported the use of therapeutic blood exchange for the treatment of degenerative diseases in older individuals. The first objective of this article is to explore the rejuvenating properties of blood-based therapies in the ageing brains and their therapeutic effects on AD. Then, we also look into the clinical applications, various limitations, and challenges associated with blood-based therapies for AD patients.
Collapse
Affiliation(s)
- Jia Yee Lee
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Mervyn Chen Xi Lim
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Rhun Yian Koh
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Min Tze Tsen
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Soi Moi Chye
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
7
|
Souza-Silva NG, Rosa DV, de Paula JJ, Coimbra RS, Miranda DM, Romano-Silva MA. Follow-up of cognitive impairment and inflammatory profile in individuals with mild COVID-19. J Neuroimmunol 2024; 389:578327. [PMID: 38489978 DOI: 10.1016/j.jneuroim.2024.578327] [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: 01/01/2024] [Revised: 02/21/2024] [Accepted: 03/03/2024] [Indexed: 03/17/2024]
Abstract
Individuals who experience mild COVID-19 can suffer from long-lasting cognitive symptoms. Notably, 26% of these individuals experience difficulties with visuospatial abilities six months after infection. However, among those who initially exhibited visuoconstructive impairments, 66% showed improvement or complete reversal over time. Additionally, changes in cytokine levels, particularly CCL11, HGF, and CXCL10, were observed. These results suggest a potential link between ongoing cognitive issues and elevated levels of pro-inflammatory cytokines, which merits further investigation.
Collapse
Affiliation(s)
- Nathália Gualberto Souza-Silva
- Centro de Tecnologia em Medicina Molecular (CTMM), Universidade Federal de Minas Gerais (UFMG), Av Alfredo Balena 190, Belo Horizonte, MG, Brazil
| | - Daniela Valadão Rosa
- Centro de Tecnologia em Medicina Molecular (CTMM), Universidade Federal de Minas Gerais (UFMG), Av Alfredo Balena 190, Belo Horizonte, MG, Brazil
| | - Jonas Jardim de Paula
- Centro de Tecnologia em Medicina Molecular (CTMM), Universidade Federal de Minas Gerais (UFMG), Av Alfredo Balena 190, Belo Horizonte, MG, Brazil; Departamento de Psiquiatria, Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Roney Santos Coimbra
- Neurogenômica/Imunopatologia, Instituto René Rachou, Fiocruz, Belo Horizonte, MG, Brazil
| | - Débora Marques Miranda
- Centro de Tecnologia em Medicina Molecular (CTMM), Universidade Federal de Minas Gerais (UFMG), Av Alfredo Balena 190, Belo Horizonte, MG, Brazil; Departamento de Pediatria, Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Marco Aurélio Romano-Silva
- Centro de Tecnologia em Medicina Molecular (CTMM), Universidade Federal de Minas Gerais (UFMG), Av Alfredo Balena 190, Belo Horizonte, MG, Brazil; Departamento de Psiquiatria, Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.
| |
Collapse
|
8
|
Aso Y, Iijima T, Jojima T, Saito M, Tanuma D, Kase M, Sakurai S, Tomaru T, Usui I. High serum levels of CCL11/Eotaxin-1 are associated with diabetic sensorimotor polyneuropathy and peripheral nerve function but not with cardiac autonomic neuropathy in people with type 2 diabetes. Postgrad Med 2024; 136:318-324. [PMID: 38660919 DOI: 10.1080/00325481.2024.2347196] [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: 11/21/2023] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
AIMS To investigate whether higher serum CCL11/Eotaxin-1, a biomarker for aging and neurodegenerative and neuroinflammatory disorders, is associated with diabetic sensorimotor polyneuropathy (DSPN), peripheral nerve dysfunction, and cardiac autonomic neuropathy in people with type 2 diabetes. METHODS This cross-sectional study included 106 patients with type 2 diabetes and 40 healthy controls, matched for the age and sex distribution of the diabetes group as a whole. The CC chemokines CCL11/Eotaxin-1 and CCL22/MDC were measured in fasting serum samples. DSPN and peripheral nerve function were assessed by neurological examination and nerve conduction studies, and cardiac autonomic function, by heart rate variability (HRV) and corrected QT (QTc) time. The cardio-ankle vascular index (CAVI) was measured as a marker for arterial stiffness. RESULTS Serum CCL11/Eotaxin-1 levels were significantly higher in diabetic patients than in healthy controls (183 ± 63.5 vs. 113.1 ± 38.5 pg/ml, p < 0.001), but serum CCL22/MDC levels were not significantly different between the two groups. In the diabetes group, the serum CCL11/Eotaxin-1 level was positively correlated with ulnar and sural nerve conduction velocities (p = 0.0009, p = 0.0208, respectively) and sensory nerve action potential (p = 0.0083), and CAVI (p = 0.0005), but not with HRV indices or QTc time, and serum CCL22/MDC was not significantly correlated with any indices of nerve conduction. In a model adjusted for age and duration of diabetes, serum CCL11/Eotaxin-1 was still associated with ulnar nerve conduction velocity (p = 0.02124). Serum CCL11/Eotaxin-1, but not CCL22/MDC, was significantly higher in patients with than in those without DSPN (208.2 ± 71.6 vs. 159.1 ± 45.1 pg/ml, respectively; p < 0.0001). CONCLUSIONS Serum CCL11/Eotaxin-1 is elevated in patients with DSPN and is associated with peripheral nerve dysfunction, in particular sensory nerve conduction velocity, suggesting that serum CCL11/Eotaxin-1 may be a potential biomarker for DSPN. CLINICAL TRIAL REGISTRATION University Hospital Medical Information Network (UMIN) Clinical Trials Registry (UMIN000040631).
Collapse
Affiliation(s)
- Yoshimasa Aso
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Toshie Iijima
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Teruo Jojima
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Masahiro Saito
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Dai Tanuma
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Masato Kase
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Shintaro Sakurai
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Takuya Tomaru
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Isao Usui
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Mibu, Tochigi, Japan
| |
Collapse
|
9
|
Garro-Núñez D, Picado-Martínez MJ, Espinoza-Campos E, Ugalde-Araya D, Macaya G, Raventós H, Chavarría-Soley G. Systematic exploration of a decade of publications on psychiatric genetics in Latin America. Am J Med Genet B Neuropsychiatr Genet 2024; 195:e32960. [PMID: 37860990 DOI: 10.1002/ajmg.b.32960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 08/08/2023] [Accepted: 09/29/2023] [Indexed: 10/21/2023]
Abstract
Psychiatric disorders have a great impact in terms of mortality, morbidity, and disability across the lifespan. Considerable effort has been devoted to understanding their complex and heterogeneous genetic architecture, including diverse ancestry populations. Our aim was to review the psychiatric genetics research published with Latin American populations from 2010 to 2019, and classify it according to country of origin, type of analysis, source of funding, and other variables. We found that most publications came from Brazil, Mexico, and Colombia. Also, local funds are generally not large enough for genome-wide studies in Latin America, with the exception of Brazil and Mexico; larger studies are often done in collaboration with international partners, mostly funded by US agencies. In most of the larger studies, the participants are individuals of Latin American ancestry living in the United States, which limits the potential for exploring the complex gene-environment interaction. Family studies, traditionally strong in Latin America, represent about 30% of the total research publications. Scarce local resources for research in Latin America have probably been an important limitation for conducting bigger and more complex studies, contributing to the reduced representation of these populations in global psychiatric genetics studies. Increasing diversity must be a goal to improve generalizability and applicability in clinical settings.
Collapse
Affiliation(s)
| | | | | | - Daniela Ugalde-Araya
- Center for Research in Cellular and Molecular Biology, Universidad de Costa Rica, San José, Costa Rica
| | - Gabriel Macaya
- Center for Research in Cellular and Molecular Biology, Universidad de Costa Rica, San José, Costa Rica
| | - Henriette Raventós
- Biology School, Universidad de Costa Rica, San José, Costa Rica
- Center for Research in Cellular and Molecular Biology, Universidad de Costa Rica, San José, Costa Rica
| | - Gabriela Chavarría-Soley
- Biology School, Universidad de Costa Rica, San José, Costa Rica
- Center for Research in Cellular and Molecular Biology, Universidad de Costa Rica, San José, Costa Rica
| |
Collapse
|
10
|
Nemes S, Logan PE, Manchella MK, Mundada NS, Joie RL, Polsinelli AJ, Hammers DB, Koeppe RA, Foroud TM, Nudelman KN, Eloyan A, Iaccarino L, Dorsant-Ardón V, Taurone A, Maryanne Thangarajah, Dage JL, Aisen P, Grinberg LT, Jack CR, Kramer J, Kukull WA, Murray ME, Rumbaugh M, Soleimani-Meigooni DN, Toga A, Touroutoglou A, Vemuri P, Atri A, Day GS, Duara R, Graff-Radford NR, Honig LS, Jones DT, Masdeu J, Mendez MF, Musiek E, Onyike CU, Riddle M, Rogalski E, Salloway S, Sha SJ, Turner RS, Wingo TS, Womack KB, Wolk DA, Rabinovici GD, Carrillo MC, Dickerson BC, Apostolova LG. Sex and APOE ε4 carrier effects on atrophy, amyloid PET, and tau PET burden in early-onset Alzheimer's disease. Alzheimers Dement 2023; 19 Suppl 9:S49-S63. [PMID: 37496307 PMCID: PMC10811272 DOI: 10.1002/alz.13403] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/28/2023]
Abstract
INTRODUCTION We used sex and apolipoprotein E ε4 (APOE ε4) carrier status as predictors of pathologic burden in early-onset Alzheimer's disease (EOAD). METHODS We included baseline data from 77 cognitively normal (CN), 230 EOAD, and 70 EO non-Alzheimer's disease (EOnonAD) participants from the Longitudinal Early-Onset Alzheimer's Disease Study (LEADS). We stratified each diagnostic group by males and females, then further subdivided each sex by APOE ε4 carrier status and compared imaging biomarkers in each stratification. Voxel-wise multiple linear regressions yielded statistical brain maps of gray matter density, amyloid, and tau PET burden. RESULTS EOAD females had greater amyloid and tau PET burdens than males. EOAD female APOE ε4 non-carriers had greater amyloid PET burdens and greater gray matter atrophy than female ε4 carriers. EOnonAD female ε4 non-carriers also had greater gray matter atrophy than female ε4 carriers. DISCUSSION The effects of sex and APOE ε4 must be considered when studying these populations. HIGHLIGHTS Novel analysis examining the effects of biological sex and apolipoprotein E ε4 (APOE ε4) carrier status on neuroimaging biomarkers among early-onset Alzheimer's disease (EOAD), early-onset non-AD (EOnonAD), and cognitively normal (CN) participants. Female sex is associated with greater pathology burden in the EOAD cohort compared to male sex. The effect of APOE ε4 carrier status on pathology burden was the most impactful in females across all cohorts.
Collapse
Affiliation(s)
- Sára Nemes
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA
| | - Paige E. Logan
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA
| | - Mohit K. Manchella
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA
- Department of Chemistry, University of Southern Indiana, Evansville, Indiana, 47712, USA
| | - Nidhi S. Mundada
- Department of Neurology, University of California, San Francisco, California, 94158, USA
| | - Renaud La Joie
- Department of Neurology, University of California, San Francisco, California, 94158, USA
| | - Angelina J. Polsinelli
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indianapolis, Indiana, 46202 USA
| | - Dustin B. Hammers
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA
| | - Robert A. Koeppe
- Department of Radiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Tatiana M. Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA
| | - Kelly N. Nudelman
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA
| | - Ani Eloyan
- Department of Biostatistics, Center for Statistical Sciences, Brown University, Providence, RI, 02912, USA
| | - Leonardo Iaccarino
- Department of Neurology, University of California, San Francisco, California, 94158, USA
| | - Valérie Dorsant-Ardón
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA
| | - Alexander Taurone
- Department of Biostatistics, Center for Statistical Sciences, Brown University, Providence, RI, 02912, USA
| | - Maryanne Thangarajah
- Department of Biostatistics, Center for Statistical Sciences, Brown University, Providence, RI, 02912, USA
| | - Jeffery L. Dage
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA
| | - Paul Aisen
- Alzheimer’s Therapeutic Research Institute, University of Southern California, San Diego, CA, 92121, USA
| | - Lea T. Grinberg
- Department of Neurology, University of California, San Francisco, California, 94158, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Clifford R. Jack
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, 55905, USA
| | - Joel Kramer
- Department of Neurology, University of California, San Francisco, California, 94158, USA
| | - Walter A. Kukull
- Department of Epidemiology, University of Washington, Seattle, Washington, USA, 98195, USA
| | - Melissa E. Murray
- Department of Neurology, Mayo Clinic, Jacksonville, Florida, 32224, USA
| | - Malia Rumbaugh
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA
| | | | - Arthur Toga
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, Los Angeles, California, 90033, USA
| | - Alexandra Touroutoglou
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Prashanthi Vemuri
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, 55905, USA
| | - Alireza Atri
- Banner Sun Health Research Institute, Sun City, Arizona, 85315, USA
| | - Gregory S. Day
- Department of Neurology, Mayo Clinic, Jacksonville, Florida, 32224, USA
| | - Ranjan Duara
- Department of Neurology, Center for Mind/Brain Medicine, Brigham & Women’s Hospital & Harvard Medical School, Boston, Massachusetts, 02115, USA
- Wein Center for Alzheimer’s Disease and Memory Disorders, Mount Sinai Medical Center, Miami, FL, 33140, USA
| | | | - Lawrence S. Honig
- Taub Institute and Department of Neurology, Columbia University Irving Medical Center, New York, New York, 10032, USA
| | - David T. Jones
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, 55905, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, 559095, USA
| | - Joseph Masdeu
- Nantz National Alzheimer Center, Houston Methodist and Weill Cornell Medicine, Houston, Texas, 77030, USA
| | - Mario F. Mendez
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, 90095, USA
| | - Erik Musiek
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, 63110, USA
| | - Chiadi U. Onyike
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Meghan Riddle
- Department of Neurology, Alpert Medical School, Brown University, Providence, Rhode Island, 02906, USA
| | - Emily Rogalski
- Department of Psychiatry and Behavioral Sciences, Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611, USA
| | - Stephen Salloway
- Department of Neurology, Alpert Medical School, Brown University, Providence, Rhode Island, 02906, USA
| | - Sharon J. Sha
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, 94304, USA
| | - Raymond S. Turner
- Department of Neurology, Georgetown Universit, Washington, DC, 20007, USA
| | - Thomas S. Wingo
- Department of Neurology and Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Kyle B. Womack
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, 63110, USA
| | - David A. Wolk
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA,19104, USA
| | - Gil D. Rabinovici
- Department of Neurology, University of California, San Francisco, California, 94158, USA
| | - Maria C. Carrillo
- Medical & Scientific Relations Division, Alzheimer’s Association, Chicago, Illinois, 60603, USA
| | - Bradford C. Dickerson
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Liana G. Apostolova
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indianapolis, Indiana, 46202 USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA
| | | |
Collapse
|
11
|
Zhao HQ, Jiang J. Chemokines and receptors in the development and progression of malignant tumors. Cytokine 2023; 170:156335. [PMID: 37591136 DOI: 10.1016/j.cyto.2023.156335] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 08/19/2023]
Abstract
Cancer cells, endothelial cells, inflammatory cells and various cytokines form a part of the tumor microenvironment (TME). Chemokines constitute the largest family of cytokines, and are mainly secreted by tumor cells and inflammatory cells in the TME. They play an important role in tumor development and progression by promoting tumor growth and metastasis, angiogenesis, and targeting the chemoattraction of inflammatory cells. Currently, some chemokine receptor antagonists are being used in clinical trials as targeted anti-tumor drugs. In this article, we review the roles of chemokines in the development and progression of malignant tumors based on recently published papers, taking into consideration of the new anti-tumor therapeutic strategies targeting chemokines and receptors.
Collapse
Affiliation(s)
- Han-Qing Zhao
- Department of General Surgery (Thyroid Surgery), Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, PR China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, PR China.
| |
Collapse
|
12
|
Cochran JN, Acosta-Uribe J, Esposito BT, Madrigal L, Aguillón D, Giraldo MM, Taylor JW, Bradley J, Fulton-Howard B, Andrews SJ, Acosta-Baena N, Alzate D, Garcia GP, Piedrahita F, Lopez HE, Anderson AG, Rodriguez-Nunez I, Roberts K, Dominantly Inherited Alzheimer Network, Absher D, Myers RM, Beecham GW, Reitz C, Rizzardi LF, Fernandez MV, Goate AM, Cruchaga C, Renton AE, Lopera F, Kosik KS. Genetic associations with age at dementia onset in the PSEN1 E280A Colombian kindred. Alzheimers Dement 2023; 19:3835-3847. [PMID: 36951251 PMCID: PMC10514237 DOI: 10.1002/alz.13021] [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: 10/31/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 03/24/2023]
Abstract
INTRODUCTION Genetic associations with Alzheimer's disease (AD) age at onset (AAO) could reveal genetic variants with therapeutic applications. We present a large Colombian kindred with autosomal dominant AD (ADAD) as a unique opportunity to discover AAO genetic associations. METHODS A genetic association study was conducted to examine ADAD AAO in 340 individuals with the PSEN1 E280A mutation via TOPMed array imputation. Replication was assessed in two ADAD cohorts, one sporadic early-onset AD study and four late-onset AD studies. RESULTS 13 variants had p<1×10-7 or p<1×10-5 with replication including three independent loci with candidate associations with clusterin including near CLU. Other suggestive associations were identified in or near HS3ST1, HSPG2, ACE, LRP1B, TSPAN10, and TSPAN14. DISCUSSION Variants with suggestive associations with AAO were associated with biological processes including clusterin, heparin sulfate, and amyloid processing. The detection of these effects in the presence of a strong mutation for ADAD reinforces their potentially impactful role.
Collapse
Affiliation(s)
| | - Juliana Acosta-Uribe
- Neuroscience Research Institute, University of California, Santa Barbara, California, and Department of Molecular Cellular and Developmental Biology University of California, Santa Barbara, California, USA
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Bianca T Esposito
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lucia Madrigal
- 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
| | - Margarita M Giraldo
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Jared W Taylor
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Joseph Bradley
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Brian Fulton-Howard
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shea J Andrews
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Natalia Acosta-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
| | - Gloria P Garcia
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Francisco Piedrahita
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Hugo E Lopez
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | | | | | - Kevin Roberts
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | | | - Devin Absher
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Richard M Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Gary W Beecham
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, USA
| | - Christiane Reitz
- Department of Epidemiology, Sergievsky Center, Taub Institute for Research on the Aging Brain, Columbia University, New York, New York, USA
| | | | | | - Alison M Goate
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Carlos Cruchaga
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alan E Renton
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Kenneth S Kosik
- Neuroscience Research Institute, University of California, Santa Barbara, California, and Department of Molecular Cellular and Developmental Biology University of California, Santa Barbara, California, USA
| |
Collapse
|
13
|
Abramova O, Zorkina Y, Ushakova V, Gryadunov D, Ikonnikova A, Fedoseeva E, Emelyanova M, Ochneva A, Morozova I, Pavlov K, Syunyakov T, Andryushchenko A, Savilov V, Kurmishev M, Andreuyk D, Shport S, Gurina O, Chekhonin V, Kostyuk G, Morozova A. Alteration of Blood Immune Biomarkers in MCI Patients with Different APOE Genotypes after Cognitive Training: A 1 Year Follow-Up Cohort Study. Int J Mol Sci 2023; 24:13395. [PMID: 37686198 PMCID: PMC10488004 DOI: 10.3390/ijms241713395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Many studies aim to detect the early phase of dementia. One of the major ways to achieve this is to identify corresponding biomarkers, particularly immune blood biomarkers. The objective of this study was to identify such biomarkers in patients with mild cognitive impairment (MCI) in an experiment that included cognitive training. A group of patients with MCI diagnoses over the age of 65 participated in the study (n = 136). Measurements of cognitive functions (using the Mini-Mental State Examination scale and Montreal Cognitive Assessment) and determination of 27 serum biomarkers were performed twice: on the first visit and on the second visit, one year after the cognitive training. APOE genotypes were also determined. Concentrations of EGF (F = 17; p = 0.00007), Eotaxin (F = 7.17; p = 0.008), GRO (F = 13.42; p = 0.0004), IL-8 (F = 8.16; p = 0.005), MCP-1 (F = 13.46; p = 0.0001) and MDC (F = 5.93; p = 0.016) increased after the cognitive training in MCI patients. All these parameters except IL-8 demonstrated a weak correlation with other immune parameters and were poorly represented in the principal component analysis. Differences in concentrations of IP-10, FGF-2, TGFa and VEGF in patients with MCI were associated with APOE genotype. Therefore, the study identified several immune blood biomarkers that could potentially be associated with changes in cognitive function.
Collapse
Affiliation(s)
- Olga Abramova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Yana Zorkina
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Valeriya Ushakova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
- Biological Faculty, M.V. Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Dmitry Gryadunov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anna Ikonnikova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Elena Fedoseeva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Marina Emelyanova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Aleksandra Ochneva
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Irina Morozova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
| | - Konstantin Pavlov
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Timur Syunyakov
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
- International Centre for Education and Research in Neuropsychiatry (ICERN), Samara State Medical University, 443016 Samara, Russia
| | - Alisa Andryushchenko
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
| | - Victor Savilov
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
| | - Marat Kurmishev
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
| | - Denis Andreuyk
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
- Biological Faculty, M.V. Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Svetlana Shport
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Olga Gurina
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Vladimir Chekhonin
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Georgy Kostyuk
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
- Department of Psychiatry, Federal State Budgetary Educational Institution of Higher Education “Moscow State University of Food Production”, Volokolamskoye Highway 11, 125080 Moscow, Russia
| | - Anna Morozova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| |
Collapse
|
14
|
Ramos J, Caywood LJ, Prough MB, Clouse JE, Herington SD, Slifer SH, Fuzzell MD, Fuzzell SL, Hochstetler SD, Miskimen KL, Main LR, Osterman MD, Zaman AF, Whitehead PL, Adams LD, Laux RA, Song YE, Foroud TM, Mayeux RP, George-Hyslop PS, Ogrocki PK, Lerner AJ, Vance JM, Cuccaro ML, Haines JL, Pericak-Vance MA, Scott WK. Genetic variants in the SHISA6 gene are associated with delayed cognitive impairment in two family datasets. Alzheimers Dement 2023; 19:611-620. [PMID: 35490390 PMCID: PMC9622429 DOI: 10.1002/alz.12686] [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: 09/23/2021] [Revised: 03/08/2022] [Accepted: 03/28/2022] [Indexed: 11/12/2022]
Abstract
INTRODUCTION Studies of cognitive impairment (CI) in Amish communities have identified sibships containing CI and cognitively unimpaired (CU) individuals. We hypothesize that CU individuals may carry protective alleles delaying age at onset (AAO) of CI. METHODS A total of 1522 individuals screened for CI were genotyped. The outcome studied was AAO for CI individuals or age at last normal exam for CU individuals. Cox mixed-effects models examined association between age and single nucleotide variants (SNVs). RESULTS Three SNVs were significantly associated (P < 5 × 10-8 ) with AAO on chromosomes 6 (rs14538074; hazard ratio [HR] = 3.35), 9 (rs534551495; HR = 2.82), and 17 (rs146729640; HR = 6.38). The chromosome 17 association was replicated in the independent National Institute on Aging Genetics Initiative for Late-Onset Alzheimer's Disease dataset. DISCUSSION The replicated genome-wide significant association with AAO on chromosome 17 is located in the SHISA6 gene, which is involved in post-synaptic transmission in the hippocampus and is a biologically plausible candidate gene for Alzheimer's disease.
Collapse
Affiliation(s)
- Jairo Ramos
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Laura J. Caywood
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michael B. Prough
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jason E. Clouse
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sharlene D. Herington
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Susan H. Slifer
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - M. Denise Fuzzell
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sarada L. Fuzzell
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | | | - Leighanne R. Main
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Michael D. Osterman
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Andrew F. Zaman
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Patrice L. Whitehead
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Larry D. Adams
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Renee A. Laux
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Yeunjoo E. Song
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Tatiana M. Foroud
- Indiana Alzheimer’s Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Richard P. Mayeux
- Taub Institute on Alzheimer’s Disease and the Aging Brain, Department of Neurology, Columbia University, New York, NY, USA
- Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
| | | | - Paula K. Ogrocki
- University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Alan J. Lerner
- University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Jeffery M. Vance
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michael L. Cuccaro
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jonathan L. Haines
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Margaret A. Pericak-Vance
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - William K. Scott
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
15
|
Li X, Zhang DF, Bi R, Tan LW, Chen X, Xu M, Yao YG. Convergent transcriptomic and genomic evidence supporting a dysregulation of CXCL16 and CCL5 in Alzheimer's disease. Alzheimers Res Ther 2023; 15:17. [PMID: 36670424 PMCID: PMC9863145 DOI: 10.1186/s13195-022-01159-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/29/2022] [Indexed: 01/22/2023]
Abstract
BACKGROUND Neuroinflammatory factors, especially chemokines, have been widely reported to be involved in the pathogenesis of Alzheimer's disease (AD). It is unclear how chemokines are altered in AD, and whether dysregulation of chemokines is the cause, or the consequence, of the disease. METHODS We initially screened the transcriptomic profiles of chemokines from publicly available datasets of brain tissues of AD patients and mouse models. Expression alteration of chemokines in the blood from AD patients was also measured to explore whether any chemokine might be used as a potential biomarker for AD. We further analyzed the association between the coding variants of chemokine genes and genetic susceptibility of AD by targeted sequencing of a Han Chinese case-control cohort. Mendelian randomization (MR) was performed to infer the causal association of chemokine dysregulation with AD development. RESULTS Three chemokine genes (CCL5, CXCL1, and CXCL16) were consistently upregulated in brain tissues from AD patients and the mouse models and were positively correlated with Aβ and tau pathology in AD mice. Peripheral blood mRNA expression of CXCL16 was upregulated in mild cognitive impairment (MCI) and AD patients, indicating the potential of CXCL16 as a biomarker for AD development. None of the coding variants within any chemokine gene conferred a genetic risk to AD. MR analysis confirmed a causal role of CCL5 dysregulation in AD mediated by trans-regulatory variants. CONCLUSIONS In summary, we have provided transcriptomic and genomic evidence supporting an active role of dysregulated CXCL16 and CCL5 during AD development.
Collapse
Affiliation(s)
- Xiao Li
- grid.419010.d0000 0004 1792 7072Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204 Yunnan China ,grid.410726.60000 0004 1797 8419Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204 China
| | - Deng-Feng Zhang
- grid.419010.d0000 0004 1792 7072Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204 Yunnan China ,grid.410726.60000 0004 1797 8419Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204 China
| | - Rui Bi
- grid.419010.d0000 0004 1792 7072Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204 Yunnan China ,grid.410726.60000 0004 1797 8419Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204 China ,grid.9227.e0000000119573309CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Li-Wen Tan
- grid.216417.70000 0001 0379 7164Mental Health Institute of the Second Xiangya Hospital, Central South University, Changsha, 410011 China
| | - Xiaogang Chen
- grid.216417.70000 0001 0379 7164Mental Health Institute of the Second Xiangya Hospital, Central South University, Changsha, 410011 China
| | - Min Xu
- grid.419010.d0000 0004 1792 7072Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204 Yunnan China ,grid.410726.60000 0004 1797 8419Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204 China
| | - Yong-Gang Yao
- grid.419010.d0000 0004 1792 7072Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204 Yunnan China ,grid.410726.60000 0004 1797 8419Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204 China ,grid.9227.e0000000119573309CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031 China
| |
Collapse
|
16
|
Markov NT, Lindbergh CA, Staffaroni AM, Perez K, Stevens M, Nguyen K, Murad NF, Fonseca C, Campisi J, Kramer J, Furman D. Age-related brain atrophy is not a homogenous process: Different functional brain networks associate differentially with aging and blood factors. Proc Natl Acad Sci U S A 2022; 119:e2207181119. [PMID: 36459652 PMCID: PMC9894212 DOI: 10.1073/pnas.2207181119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 10/04/2022] [Indexed: 12/04/2022] Open
Abstract
Aging is characterized by a progressive loss of brain volume at an estimated rate of 5% per decade after age 40. While these morphometric changes, especially those affecting gray matter and atrophy of the temporal lobe, are predictors of cognitive performance, the strong association with aging obscures the potential parallel, but more specific role, of individual subject physiology. Here, we studied a cohort of 554 human subjects who were monitored using structural MRI scans and blood immune protein concentrations. Using machine learning, we derived a cytokine clock (CyClo), which predicted age with good accuracy (Mean Absolute Error = 6 y) based on the expression of a subset of immune proteins. These proteins included, among others, Placenta Growth Factor (PLGF) and Vascular Endothelial Growth Factor (VEGF), both involved in angiogenesis, the chemoattractant vascular cell adhesion molecule 1 (VCAM-1), the canonical inflammatory proteins interleukin-6 (IL-6) and tumor necrosis factor alpha (TNFα), the chemoattractant IP-10 (CXCL10), and eotaxin-1 (CCL11), previously involved in brain disorders. Age, sex, and the CyClo were independently associated with different functionally defined cortical networks in the brain. While age was mostly correlated with changes in the somatomotor system, sex was associated with variability in the frontoparietal, ventral attention, and visual networks. Significant canonical correlation was observed for the CyClo and the default mode, limbic, and dorsal attention networks, indicating that immune circulating proteins preferentially affect brain processes such as focused attention, emotion, memory, response to social stress, internal evaluation, and access to consciousness. Thus, we identified immune biomarkers of brain aging which could be potential therapeutic targets for the prevention of age-related cognitive decline.
Collapse
Affiliation(s)
- Nikola T. Markov
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA94945
| | - Cutter A. Lindbergh
- Department of Neurology, Memory and Aging Center, University of California San Francisco, Weill Institute for Neurosciences, San Francisco, CA94158
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT06030
| | - Adam M. Staffaroni
- Department of Neurology, Memory and Aging Center, University of California San Francisco, Weill Institute for Neurosciences, San Francisco, CA94158
| | - Kevin Perez
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA94945
- University of Lausanne, LausanneCH-1015, Switzerland
| | - Michael Stevens
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA94945
| | - Khiem Nguyen
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA94945
- Nguyen Tat Thanh Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City70000, Vietnam
| | - Natalia F. Murad
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA94945
| | - Corrina Fonseca
- Department of Neurology, Memory and Aging Center, University of California San Francisco, Weill Institute for Neurosciences, San Francisco, CA94158
| | - Judith Campisi
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA94945
| | - Joel Kramer
- Department of Neurology, Memory and Aging Center, University of California San Francisco, Weill Institute for Neurosciences, San Francisco, CA94158
| | - David Furman
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA94945
- Instituto de Investigaciones en Medicina Traslacional, Universidad Austral, Consejo Nacional de Investigaciones Científicas y Técnicas, Pilar1629, Argentina
- Stanford 1000 Immunomes Project, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
17
|
Aerqin Q, Wang ZT, Wu KM, He XY, Dong Q, Yu JT. Omics-based biomarkers discovery for Alzheimer's disease. Cell Mol Life Sci 2022; 79:585. [PMID: 36348101 PMCID: PMC11803048 DOI: 10.1007/s00018-022-04614-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 10/22/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorders presenting with the pathological hallmarks of amyloid plaques and tau tangles. Over the past few years, great efforts have been made to explore reliable biomarkers of AD. High-throughput omics are a technology driven by multiple levels of unbiased data to detect the complex etiology of AD, and it provides us with new opportunities to better understand the pathophysiology of AD and thereby identify potential biomarkers. Through revealing the interaction networks between different molecular levels, the ultimate goal of multi-omics is to improve the diagnosis and treatment of AD. In this review, based on the current AD pathology and the current status of AD diagnostic biomarkers, we summarize how genomics, transcriptomics, proteomics and metabolomics are all conducing to the discovery of reliable AD biomarkers that could be developed and used in clinical AD management.
Collapse
Affiliation(s)
- Qiaolifan Aerqin
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Kai-Min Wu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Xiao-Yu He
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
18
|
Peng W, Xie Y, Liao C, Bai Y, Wang H, Li C. Spatiotemporal patterns of gliosis and neuroinflammation in presenilin 1/2 conditional double knockout mice. Front Aging Neurosci 2022; 14:966153. [PMID: 36185485 PMCID: PMC9521545 DOI: 10.3389/fnagi.2022.966153] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022] Open
Abstract
Increasing evidence indicates that neuroinflammation contributes to and exacerbates the pathogenesis of Alzheimer’s disease (AD). Neuroinflammation is thought to be primarily driven by glial cells (microglia and astrocytes) and escalates with neurodegenerative progression in AD. However, the spatiotemporal change patterns of glial reactivity and neuroinflammatory response during different stages of neurodegeneration, especially early in disease, remain unknown. Here we found that gliosis and the up-regulation of substantial neuroinflammatory genes were primarily initiated in the cortex of presenilin 1/2 conditional double knockout (cDKO) mice, rather than in the hippocampus. Specifically, astrocyte activation preceding microglial activation was found in the somatosensory cortex (SS) of cDKO mice at 6 weeks of age. Over time, both astrocyte and microglial activation were found in the whole cortex, and age-related increases in gliosis activation were more pronounced in the cortex compared to hippocampus. Moreover, the age-associated increase in glial activation was accompanied by a gradual increase in the expression of cell chemokines Ccl3 and Ccl4, complement related factors C1qb, C3 and C4, and lysosomal proteases cathepsin S and Z. These findings suggest that astrocyte and microglial activation with a concurrent increase in inflammatory mediators such as chemokines might be an early event and contribute to the pathogenesis of neurodegeneration due to presenilin deficiency.
Collapse
Affiliation(s)
- Wenjun Peng
- Key Laboratory of Brain Functional Genomics (STCSM and MOE), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai, China
| | - Yuan Xie
- Key Laboratory of Brain Functional Genomics (STCSM and MOE), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai, China
| | - Chongzheng Liao
- Key Laboratory of Brain Functional Genomics (STCSM and MOE), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai, China
| | - Yunxia Bai
- Key Laboratory of Brain Functional Genomics (STCSM and MOE), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai, China
| | - Huimin Wang
- Key Laboratory of Brain Functional Genomics (STCSM and MOE), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai, China
- Shanghai Changning Mental Health Center, Shanghai, China
- NYU-ECNU Institute of Brain and Cognitive Science at NYU Shanghai, Shanghai, China
- Huimin Wang,
| | - Chunxia Li
- Key Laboratory of Brain Functional Genomics (STCSM and MOE), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai, China
- Shanghai Changning Mental Health Center, Shanghai, China
- *Correspondence: Chunxia Li,
| |
Collapse
|
19
|
Nordestgaard LT, Christoffersen M, Frikke-Schmidt R. Shared Risk Factors between Dementia and Atherosclerotic Cardiovascular Disease. Int J Mol Sci 2022; 23:9777. [PMID: 36077172 PMCID: PMC9456552 DOI: 10.3390/ijms23179777] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/21/2022] [Accepted: 08/24/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer's disease is the most common form of dementia, and the prodromal phases of Alzheimer's disease can last for decades. Vascular dementia is the second most common form of dementia and is distinguished from Alzheimer's disease by evidence of previous stroke or hemorrhage and current cerebrovascular disease. A compiled group of vascular-related dementias (vascular dementia and unspecified dementia) is often referred to as non-Alzheimer dementia. Recent evidence indicates that preventing dementia by lifestyle interventions early in life with a focus on reducing cardiovascular risk factors is a promising strategy for reducing future risk. Approximately 40% of dementia cases is estimated to be preventable by targeting modifiable, primarily cardiovascular risk factors. The aim of this review is to describe the association between risk factors for atherosclerotic cardiovascular disease and the risk of Alzheimer's disease and non-Alzheimer dementia by providing an overview of the current evidence and to shed light on possible shared pathogenic pathways between dementia and cardiovascular disease. The included risk factors are body mass index (BMI); plasma triglyceride-, high-density lipoprotein (HDL) cholesterol-, low-density lipoprotein (LDL) cholesterol-, and total cholesterol concentrations; hypertension; diabetes; non-alcoholic fatty liver disease (NAFLD); physical inactivity; smoking; diet; the gut microbiome; and genetics. Furthermore, we aim to disentangle the difference between associations of risk factors in midlife as compared with in late life.
Collapse
Affiliation(s)
- Liv Tybjærg Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark
| | - Mette Christoffersen
- Department of Clinical Biochemistry, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
20
|
Nazarinia D, Behzadifard M, Gholampour J, Karimi R, Gholampour M. Eotaxin-1 (CCL11) in neuroinflammatory disorders and possible role in COVID-19 neurologic complications. Acta Neurol Belg 2022; 122:865-869. [PMID: 35690992 PMCID: PMC9188656 DOI: 10.1007/s13760-022-01984-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 05/18/2022] [Indexed: 12/20/2022]
Abstract
The related neurologic complications of SARS-CoV-2 infection in COVID-19 patients and survivors comprise symptoms including depression, anxiety, muscle pain, dizziness, headaches, fatigue, and anosmia/hyposmia that may continue for months. Recent studies have been demonstrated that chemokines have brain-specific attraction and effects such as chemotaxis, cell adhesion, modulation of neuroendocrine functions, and neuroinflammation. CCL11 is a member of the eotaxin family that is chemotactic agents for eosinophils and participate in innate immunity. Eotaxins may exert physiological and pathological functions in the central nerve system, and CCL11 may induce neuronal cytotoxicity effects by inducing the production of reactive oxygen species (ROS) in microglia cells. Plasma levels of CCL11 elevated in neuroinflammation and neurodegenerative disorders. COVID-19 patients display elevations in CCL11 levels. As CCL11 plays roles in physiosomatic and neuroinflammation, analyzing the level of this chemokine in COVID-19 patients during hospitalization and to predicting post-COVID-19-related neurologic complications may be worthwhile. Moreover, using chemokine modulators may be helpful in lessening the neurologic complications in such patients.
Collapse
Affiliation(s)
- Donya Nazarinia
- Department of Physiology, School of Paramedical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| | - Mahin Behzadifard
- Department of Physiology, School of Paramedical Sciences, Dezful University of Medical Sciences, Dezful, Iran.
| | - Javad Gholampour
- Department of Nursing, Faculty of Nursing and Midwifery, Mashhad Branch of Islamic Azad University, Mashhad, Iran
| | - Roqaye Karimi
- Department of Hematology and Cell Therapy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammadali Gholampour
- Department of Medicine, Lung Biology Center, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| |
Collapse
|
21
|
Sirkis DW, Bonham LW, Johnson TP, La Joie R, Yokoyama JS. Dissecting the clinical heterogeneity of early-onset Alzheimer's disease. Mol Psychiatry 2022; 27:2674-2688. [PMID: 35393555 PMCID: PMC9156414 DOI: 10.1038/s41380-022-01531-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/07/2022] [Accepted: 03/16/2022] [Indexed: 12/14/2022]
Abstract
Early-onset Alzheimer's disease (EOAD) is a rare but particularly devastating form of AD. Though notable for its high degree of clinical heterogeneity, EOAD is defined by the same neuropathological hallmarks underlying the more common, late-onset form of AD. In this review, we describe the various clinical syndromes associated with EOAD, including the typical amnestic phenotype as well as atypical variants affecting visuospatial, language, executive, behavioral, and motor functions. We go on to highlight advances in fluid biomarker research and describe how molecular, structural, and functional neuroimaging can be used not only to improve EOAD diagnostic acumen but also enhance our understanding of fundamental pathobiological changes occurring years (and even decades) before the onset of symptoms. In addition, we discuss genetic variation underlying EOAD, including pathogenic variants responsible for the well-known mendelian forms of EOAD as well as variants that may increase risk for the much more common forms of EOAD that are either considered to be sporadic or lack a clear autosomal-dominant inheritance pattern. Intriguingly, specific pathogenic variants in PRNP and MAPT-genes which are more commonly associated with other neurodegenerative diseases-may provide unexpectedly important insights into the formation of AD tau pathology. Genetic analysis of the atypical clinical syndromes associated with EOAD will continue to be challenging given their rarity, but integration of fluid biomarker data, multimodal imaging, and various 'omics techniques and their application to the study of large, multicenter cohorts will enable future discoveries of fundamental mechanisms underlying the development of EOAD and its varied clinical presentations.
Collapse
Affiliation(s)
- Daniel W Sirkis
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Luke W Bonham
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Taylor P Johnson
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Jennifer S Yokoyama
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA.
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94158, USA.
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW This article discusses the spectrum of genetic risk in familial and sporadic forms of early- and late-onset Alzheimer disease (AD). Recent work illuminating the complex genetic architecture of AD is discussed in the context of high and low risk and what is known in different populations. RECENT FINDINGS A small proportion of AD is autosomal dominant familial AD caused by variants in PSEN1, PSEN2, or APP, although more recently described rare genetic changes can also increase risk substantially over the general population, with odds ratios estimated at 2 to 4. APOE remains the strongest genetic risk factor for late-onset AD, and understanding the biology of APOE has yielded mechanistic insights and leads for therapeutic interventions. Genome-wide studies enabled by rapidly developing technologic advances in sequencing have identified numerous risk factors that have a low impact on risk but are widely shared throughout the population and involve a repertoire of cell pathways, again shining light on potential paths to intervention. Population studies aimed at defining and stratifying genetic AD risk have been informative, although they are not yet widely applicable clinically because the studies were not performed in people with diverse ancestry and ethnicity and thus population-wide data are lacking. SUMMARY The value of genetic information to practitioners in the clinic is distinct from information sought by researchers looking to identify novel therapeutic targets. It is possible to envision a future in which genetic stratification joins other biomarkers to facilitate therapeutic choices and inform prognosis. Genetics already has transformed our understanding of AD pathogenesis and will, no doubt, continue to reveal the complexity of brain biology in health and disease.
Collapse
|
23
|
Xiao X, Liao X, Zhou Y, Weng L, Guo L, Zhou L, Wang X, Liu X, Liu H, Bi X, Xu T, Zhu Y, Yang Q, Zhang S, Hao X, Liu Y, Zhang W, Li J, Shen L, Jiao B. Variants in the Niemann-Pick type C genes are not associated with Alzheimer's disease: A large case-control study in the Chinese Population. Neurobiol Aging 2022; 116:49-54. [DOI: 10.1016/j.neurobiolaging.2022.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 10/18/2022]
|
24
|
Scabia G, Testa G, Scali M, Del Turco S, Desiato G, Berardi N, Sale A, Matteoli M, Maffei L, Maffei M, Mainardi M. Reduced ccl11/eotaxin mediates the beneficial effects of environmental stimulation on the aged hippocampus. Brain Behav Immun 2021; 98:234-244. [PMID: 34418501 DOI: 10.1016/j.bbi.2021.08.222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 07/15/2021] [Accepted: 08/13/2021] [Indexed: 02/08/2023] Open
Abstract
A deterioration in cognitive performance accompanies brain aging, even in the absence of neurodegenerative pathologies. However, the rate of cognitive decline can be slowed down by enhanced cognitive and sensorimotor stimulation protocols, such as environmental enrichment (EE). Understanding how EE exerts its beneficial effects on the aged brain pathophysiology can help in identifying new therapeutic targets. In this regard, the inflammatory chemokine ccl11/eotaxin-1 is a marker of aging with a strong relevance for neurodegenerative processes. Here, we demonstrate that EE in both elderly humans and aged mice decreases circulating levels of ccl11. Interfering, in mice, with the ccl11 decrease induced by EE ablated the beneficial effects on long-term memory retention, hippocampal neurogenesis, activation of local microglia and of ribosomal protein S6. On the other hand, treatment of standard-reared aged mice with an anti-ccl11 antibody resulted in EE-like improvements in spatial memory, hippocampal neurogenesis, and microglial activation. Taken together, our findings point to a decrease in circulating ccl11 concentration as a key mediator of the enhanced hippocampal function resulting from exposure to EE.
Collapse
Affiliation(s)
- Gaia Scabia
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Pisa, Italy; Obesity and Lipodystrophies Center at Pisa University Hospital, Pisa, Italy
| | - Giovanna Testa
- Laboratory of Biology "Bio@SNS", Scuola Normale Superiore, Pisa, Italy
| | - Manuela Scali
- Institute of Neuroscience, National Research Council (IN-CNR), Pisa, Italy
| | - Serena Del Turco
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Pisa, Italy
| | - Genni Desiato
- Institute of Neuroscience, National Research Council (IN-CNR), Milan, Italy; Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Nicoletta Berardi
- Institute of Neuroscience, National Research Council (IN-CNR), Pisa, Italy; Department of Neuroscience, Psychology, Drug Research and Child Health, NEUROFARBA University of Florence, Florence, Italy
| | - Alessandro Sale
- Institute of Neuroscience, National Research Council (IN-CNR), Pisa, Italy
| | - Michela Matteoli
- Institute of Neuroscience, National Research Council (IN-CNR), Milan, Italy; Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Lamberto Maffei
- Laboratory of Biology "Bio@SNS", Scuola Normale Superiore, Pisa, Italy; Institute of Neuroscience, National Research Council (IN-CNR), Pisa, Italy
| | - Margherita Maffei
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Pisa, Italy; Obesity and Lipodystrophies Center at Pisa University Hospital, Pisa, Italy.
| | - Marco Mainardi
- Laboratory of Biology "Bio@SNS", Scuola Normale Superiore, Pisa, Italy; Institute of Neuroscience, National Research Council (IN-CNR), Pisa, Italy.
| | | |
Collapse
|
25
|
Guven G, Samanci B, Gulec C, Hanagasi H, Gurvit H, Gokalp EE, Tepgec F, Guler S, Uyguner O, Bilgic B. A novel PSEN2 p.Ser175Phe variant in a family with Alzheimer's disease. Neurol Sci 2021; 42:2497-2504. [PMID: 33855622 DOI: 10.1007/s10072-021-05243-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 04/09/2021] [Indexed: 11/28/2022]
Abstract
Alzheimer's disease (AD) can be either sporadic or familial, and familial forms of AD accounts for only 5% of the cases. So far, autosomal dominantly inherited mutations in "Presenilin 1" (PSEN1), "Presenilin 2" (PSEN2), and "Amyloid precursor protein" (APP) genes were associated with familial AD. Amid the others, pathogenic mutations in the PSEN2 gene are less common. In this study, we describe a novel heterozygous PSEN2 (c.524C>T, p.Ser175Phe) alteration identified in a 58-year-old Turkish patient from a family with multiple dementia cases. This variant was further present in the patient's clinically affected maternal cousin as well as in the asymptomatic mother and two maternal aunts who were carriers of the APOE ε2/ε3 genotype. The variant is located in the conserved residue of transmembrane domain III encoded by exon 6 of the major transcript. In silico protein structure analyses predicted that this variant might change the architecture of interaction between the two alpha helixes of PSEN2. We propose that p.Ser175Phe may have a pathogenic effect on protein function and may play a significant role in the molecular pathways leading to Alzheimer's disease in this family.
Collapse
Affiliation(s)
- Gamze Guven
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey.
| | - Bedia Samanci
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Cagri Gulec
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Hasmet Hanagasi
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Hakan Gurvit
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ebru Erzurumluoglu Gokalp
- Department of Medical Genetics, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Fatih Tepgec
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Suleyman Guler
- Neurology Clinic, Mehmet Akif Inan Training and Research Hospital, University of Health Sciences, Sanliurfa, Turkey
| | - Oya Uyguner
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Basar Bilgic
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
26
|
Uddin MS, Hasana S, Hossain MF, Islam MS, Behl T, Perveen A, Hafeez A, Ashraf GM. Molecular Genetics of Early- and Late-Onset Alzheimer's Disease. Curr Gene Ther 2021; 21:43-52. [PMID: 33231156 DOI: 10.2174/1566523220666201123112822] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia in the elderly and this complex disorder is associated with environmental as well as genetic factors. Early-onset AD (EOAD) and late-onset AD (LOAD, more common) are major identified types of AD. The genetics of EOAD is extensively understood, with three gene variants such as APP, PSEN1, and PSEN2 leading to the disease. Some common alleles, including APOE, are effectively associated with LOAD identified, but the genetics of LOAD is not clear to date. It has been accounted that about 5-10% of EOAD patients can be explained through mutations in the three familiar genes of EOAD. The APOE ε4 allele augmented the severity of EOAD risk in carriers, and the APOE ε4 allele was considered as a hallmark of EOAD. A great number of EOAD patients, who are not genetically explained, indicate that it is not possible to identify disease-triggering genes yet. Although several genes have been identified by using the technology of next-generation sequencing in EOAD families, including SORL1, TYROBP, and NOTCH3. A number of TYROBP variants are identified through exome sequencing in EOAD patients and these TYROBP variants may increase the pathogenesis of EOAD. The existence of the ε4 allele is responsible for increasing the severity of EOAD. However, several ε4 allele carriers propose the presence of other LOAD genetic as well as environmental risk factors that are not identified yet. It is urgent to find out missing genetics of EOAD and LOAD etiology to discover new potential genetic facets which will assist in understanding the pathological mechanism of AD. These investigations should contribute to developing a new therapeutic candidate for alleviating, reversing and preventing AD. This article, based on current knowledge, represents the overview of the susceptible genes of EOAD, and LOAD. Next, we represent the probable molecular mechanism that might elucidate the genetic etiology of AD and highlight the role of massively parallel sequencing technologies for novel gene discoveries.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - Sharifa Hasana
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | | | | | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, India
| | - Asma Perveen
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Abdul Hafeez
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
27
|
Sepulveda-Falla D, Chavez-Gutierrez L, Portelius E, Vélez JI, Dujardin S, Barrera-Ocampo A, Dinkel F, Hagel C, Puig B, Mastronardi C, Lopera F, Hyman BT, Blennow K, Arcos-Burgos M, de Strooper B, Glatzel M. A multifactorial model of pathology for age of onset heterogeneity in familial Alzheimer's disease. Acta Neuropathol 2021; 141:217-233. [PMID: 33319314 PMCID: PMC7847436 DOI: 10.1007/s00401-020-02249-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/22/2020] [Accepted: 11/23/2020] [Indexed: 12/13/2022]
Abstract
Presenilin-1 (PSEN1) mutations cause familial Alzheimer's disease (FAD) characterized by early age of onset (AoO). Examination of a large kindred harboring the PSEN1-E280A mutation reveals a range of AoO spanning 30 years. The pathophysiological drivers and clinical impact of AoO variation in this population are unknown. We examined brains of 23 patients focusing on generation and deposition of beta-amyloid (Aβ) and Tau pathology profile. In 14 patients distributed at the extremes of AoO, we performed whole-exome capture to identify genotype-phenotype correlations. We also studied kinome activity, proteasome activity, and protein polyubiquitination in brain tissue, associating it with Tau phosphorylation profiles. PSEN1-E280A patients showed a bimodal distribution for AoO. Besides AoO, there were no clinical differences between analyzed groups. Despite the effect of mutant PSEN1 on production of Aβ, there were no relevant differences between groups in generation and deposition of Aβ. However, differences were found in hyperphosphorylated Tau (pTau) pathology, where early onset patients showed severe pathology with diffuse aggregation pattern associated with increased activation of stress kinases. In contrast, late-onset patients showed lesser pTau pathology and a distinctive kinase activity. Furthermore, we identified new protective genetic variants affecting ubiquitin-proteasome function in early onset patients, resulting in higher ubiquitin-dependent degradation of differentially phosphorylated Tau. In PSEN1-E280A carriers, altered γ-secretase activity and resulting Aβ accumulation are prerequisites for early AoO. However, Tau hyperphosphorylation pattern, and its degradation by the proteasome, drastically influences disease onset in individuals with otherwise similar Aβ pathology, hinting toward a multifactorial model of disease for FAD. In sporadic AD (SAD), a wide range of heterogeneity, also influenced by Tau pathology, has been identified. Thus, Tau-induced heterogeneity is a common feature in both AD variants, suggesting that a multi-target therapeutic approach should be used to treat AD.
Collapse
Affiliation(s)
- Diego Sepulveda-Falla
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Neuroscience Group of Antioquia, Faculty of Medicine, University of Antioquia, Medellín, Colombia.
| | - Lucia Chavez-Gutierrez
- VIB Center for Brain and Disease Research, 3000, Leuven, Belgium
- Department of Neurology, KU Leuven, Leuven, Belgium
| | - Erik Portelius
- Institute of Neuroscience and Physiology, Dept. of Psychiatry and Neurochemistry, The Sahlgrenska Academy At the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 431 80, Mölndal, Sweden
| | - Jorge I Vélez
- Department of Genome Sciences, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Universidad del Norte, Barranquilla, Colombia
| | - Simon Dujardin
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Charlestown, USA
| | - Alvaro Barrera-Ocampo
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Facultad de Ciencias Naturales, Departamento de Ciencias Farmaceuticas, Universidad Icesi, Grupo Natura, Calle 18 No. 122 -135, Cali, Colombia
| | - Felix Dinkel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Hagel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Berta Puig
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudio Mastronardi
- Department of Genome Sciences, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- GIPSI Group, Department of Psychiatry, Medical Research Institute, University of Antioquia, Medellín, Colombia
| | - Francisco Lopera
- Neuroscience Group of Antioquia, Faculty of Medicine, University of Antioquia, Medellín, Colombia
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Charlestown, USA
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Dept. of Psychiatry and Neurochemistry, The Sahlgrenska Academy At the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 431 80, Mölndal, Sweden
| | - Mauricio Arcos-Burgos
- GIPSI Group, Department of Psychiatry, Medical Research Institute, University of Antioquia, Medellín, Colombia
| | - Bart de Strooper
- VIB Center for Brain and Disease Research, 3000, Leuven, Belgium
- Department of Neurology, KU Leuven, Leuven, Belgium
- UK Dementia Research Institute, University College London, Queen Square, London, WC1N 3BG, UK
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
28
|
Does a hypoxic injury from a non-fatal overdose lead to an Alzheimer Disease? Neurochem Int 2020; 143:104936. [PMID: 33309980 DOI: 10.1016/j.neuint.2020.104936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/21/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022]
Abstract
Long term consequence of non-fatal overdose in people who use opioids are not well understood. The intermittent exposure to non-fatal overdose leads to a tauopathy that is often accompanied by abrogated neuroprotective response, abnormal amyloid processing and other pathologies. The scope and limitations of available literature are discussed including neuropathologies associated with opioid and overdose exposures, contributing comorbidities and proteinopathies. Contrasting postmortem data of overdose victims with animal models of opioid neuropathologies and hypoxic injury paints a picture distinct from other proteinopathies as well as effects of moderate opioid exposure. Furthermore the reported biochemical changes and potential targets for therapeutic intervention were mapped pointing to underlying imbalance between tau kinases and phosphatases that is characteristic of Alzheimer Disease.
Collapse
|
29
|
CCL-11 or Eotaxin-1: An Immune Marker for Ageing and Accelerated Ageing in Neuro-Psychiatric Disorders. Pharmaceuticals (Basel) 2020; 13:ph13090230. [PMID: 32887304 PMCID: PMC7558796 DOI: 10.3390/ph13090230] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/27/2020] [Accepted: 08/31/2020] [Indexed: 12/16/2022] Open
Abstract
Background: CCL-11 (eotaxin) is a chemokine with an important role in allergic conditions. Recent evidence indicates that CCL-11 plays a role in brain disorders as well. This paper reviews the associations between CCL-11 and aging, neurodegenerative, neuroinflammatory and neuropsychiatric disorders. Methods: Electronic databases were searched for original articles examining CCL-11 in neuropsychiatric disorders. Results: CCL-11 is rapidly transported from the blood to the brain through the blood-brain barrier. Age-related increases in CCL-11 are associated with cognitive impairments in executive functions and episodic and semantic memory, and therefore, this chemokine has been described as an “Endogenous Cognition Deteriorating Chemokine” (ECDC) or “Accelerated Brain-Aging Chemokine” (ABAC). In schizophrenia, increased CCL-11 is not only associated with impairments in cognitive functions, but also with key symptoms including formal thought disorders. Some patients with mood disorders and premenstrual syndrome show increased plasma CCL-11 levels. In diseases of old age, CCL-11 is associated with lowered neurogenesis and neurodegenerative processes, and as a consequence, increased CCL-11 increases risk towards Alzheimer’s disease. Polymorphisms in the CCL-11 gene are associated with stroke. Increased CCL-11 also plays a role in neuroinflammatory disease including multiple sclerosis. In animal models, neutralization of CCL-11 may protect against nigrostriatal neurodegeneration. Increased production of CCL-11 may be attenuated by glucocorticoids, minocycline, resveratrol and anti-CCL11 antibodies. Conclusions: Increased CCL-11 production during inflammatory conditions may play a role in human disease including age-related cognitive decline, schizophrenia, mood disorders and neurodegenerative disorders. Increased CCL-11 production is a new drug target in the treatment and prevention of those disorders.
Collapse
|
30
|
Ohnmacht J, May P, Sinkkonen L, Krüger R. Missing heritability in Parkinson's disease: the emerging role of non-coding genetic variation. J Neural Transm (Vienna) 2020; 127:729-748. [PMID: 32248367 PMCID: PMC7242266 DOI: 10.1007/s00702-020-02184-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 03/24/2020] [Indexed: 02/01/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder caused by a complex interplay of genetic and environmental factors. For the stratification of PD patients and the development of advanced clinical trials, including causative treatments, a better understanding of the underlying genetic architecture of PD is required. Despite substantial efforts, genome-wide association studies have not been able to explain most of the observed heritability. The majority of PD-associated genetic variants are located in non-coding regions of the genome. A systematic assessment of their functional role is hampered by our incomplete understanding of genotype-phenotype correlations, for example through differential regulation of gene expression. Here, the recent progress and remaining challenges for the elucidation of the role of non-coding genetic variants is reviewed with a focus on PD as a complex disease with multifactorial origins. The function of gene regulatory elements and the impact of non-coding variants on them, and the means to map these elements on a genome-wide level, will be delineated. Moreover, examples of how the integration of functional genomic annotations can serve to identify disease-associated pathways and to prioritize disease- and cell type-specific regulatory variants will be given. Finally, strategies for functional validation and considerations for suitable model systems are outlined. Together this emphasizes the contribution of rare and common genetic variants to the complex pathogenesis of PD and points to remaining challenges for the dissection of genetic complexity that may allow for better stratification, improved diagnostics and more targeted treatments for PD in the future.
Collapse
Affiliation(s)
- Jochen Ohnmacht
- LCSB, University of Luxembourg, Belvaux, Luxembourg
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Belvaux, Luxembourg
| | - Patrick May
- LCSB, University of Luxembourg, Belvaux, Luxembourg
| | - Lasse Sinkkonen
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Belvaux, Luxembourg
| | - Rejko Krüger
- LCSB, University of Luxembourg, Belvaux, Luxembourg.
- Luxembourg Institute of Health (LIH), Transversal Translational Medicine, Strassen, Luxembourg.
- Parkinson Research Clinic, Centre Hospitalier de Luxembourg (CHL), Luxembourg, Luxembourg.
| |
Collapse
|
31
|
Piaceri I, Chiari A, Galli C, Bagnoli S, Ferrari C, Saavedra ST, Molinari MA, Vinceti G, Sorbi S, Nacmias B. Incomplete penetrance in familial Alzheimer’s disease with PSEN1 Ala260Gly mutation. Neurol Sci 2020; 41:2263-2266. [DOI: 10.1007/s10072-020-04421-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 04/13/2020] [Indexed: 01/06/2023]
|
32
|
The Genetics of Alzheimer's Disease in the Chinese Population. Int J Mol Sci 2020; 21:ijms21072381. [PMID: 32235595 PMCID: PMC7178026 DOI: 10.3390/ijms21072381] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/22/2020] [Accepted: 03/27/2020] [Indexed: 12/30/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease characterized by progressive cognitive dysfunction and behavioral impairment. In China, the number of AD patients is growing rapidly, which poses a considerable burden on society and families. In recent years, through the advancement of genome-wide association studies, second-generation gene sequencing technology, and their application in AD genetic research, more genetic loci associated with the risk for AD have been discovered, including KCNJ15, TREM2, and GCH1, which provides new ideas for the etiology and treatment of AD. This review summarizes three early-onset AD causative genes (APP, PSEN1, and PSEN2) and some late-onset AD susceptibility genes and their mutation sites newly discovered in China, and briefly introduces the potential mechanisms of these genetic susceptibilities in the pathogenesis of AD, which would help in understanding the genetic mechanisms underlying this devastating disease.
Collapse
|
33
|
Kuznik BI, Guseva ES, Davydov SO, Smolyakov YN, Tsybikov NN. The effects of the “youth protein” GDF11 and “aging proteins” ccL11, GDF15, JAM-A on cardiohemodynamics in women with essential hypertension. ACTA ACUST UNITED AC 2020. [DOI: 10.18705/1607-419x-2019-25-5-527-539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
|
34
|
Arboleda-Velasquez JF, Lopera F, O'Hare M, Delgado-Tirado S, Marino C, Chmielewska N, Saez-Torres KL, Amarnani D, Schultz AP, Sperling RA, Leyton-Cifuentes D, Chen K, Baena A, Aguillon D, Rios-Romenets S, Giraldo M, Guzmán-Vélez E, Norton DJ, Pardilla-Delgado E, Artola A, Sanchez JS, Acosta-Uribe J, Lalli M, Kosik KS, Huentelman MJ, Zetterberg H, Blennow K, Reiman RA, Luo J, Chen Y, Thiyyagura P, Su Y, Jun GR, Naymik M, Gai X, Bootwalla M, Ji J, Shen L, Miller JB, Kim LA, Tariot PN, Johnson KA, Reiman EM, Quiroz YT. Resistance to autosomal dominant Alzheimer's disease in an APOE3 Christchurch homozygote: a case report. Nat Med 2019; 25:1680-1683. [PMID: 31686034 PMCID: PMC6898984 DOI: 10.1038/s41591-019-0611-3] [Citation(s) in RCA: 371] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 09/12/2019] [Indexed: 02/07/2023]
Abstract
We identified a PSEN1 (presenilin 1) mutation carrier from the world's largest autosomal dominant Alzheimer's disease kindred, who did not develop mild cognitive impairment until her seventies, three decades after the expected age of clinical onset. The individual had two copies of the APOE3 Christchurch (R136S) mutation, unusually high brain amyloid levels and limited tau and neurodegenerative measurements. Our findings have implications for the role of APOE in the pathogenesis, treatment and prevention of Alzheimer's disease.
Collapse
Affiliation(s)
- Joseph F Arboleda-Velasquez
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia de la Universidad de Antioquia, Medellin, Colombia
| | - Michael O'Hare
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Santiago Delgado-Tirado
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Claudia Marino
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Natalia Chmielewska
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Boston College, Boston, MA, USA
| | - Kahira L Saez-Torres
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Dhanesh Amarnani
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Aaron P Schultz
- Massachusetts General Hospital and Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Reisa A Sperling
- Massachusetts General Hospital and Department of Neurology, Harvard Medical School, Boston, MA, USA
- Brigham and Women's Hospital and the Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - David Leyton-Cifuentes
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Universidad Escuela de Ingenieria de Antioquia-EIA, Medellin, Colombia
| | - Kewei Chen
- The Banner Alzheimer's Institute, Phoenix, AZ, USA
- University of Arizona, Tucson, AZ, USA
- Arizona State University, Tempe, AZ, USA
| | - Ana Baena
- Grupo de Neurociencias de Antioquia de la Universidad de Antioquia, Medellin, Colombia
| | - David Aguillon
- Grupo de Neurociencias de Antioquia de la Universidad de Antioquia, Medellin, Colombia
| | - Silvia Rios-Romenets
- Grupo de Neurociencias de Antioquia de la Universidad de Antioquia, Medellin, Colombia
| | - Margarita Giraldo
- Grupo de Neurociencias de Antioquia de la Universidad de Antioquia, Medellin, Colombia
| | - Edmarie Guzmán-Vélez
- Massachusetts General Hospital and the Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Daniel J Norton
- Massachusetts General Hospital and the Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Department of Psychology, Gordon College, Wenham, MA, USA
| | | | - Arabiye Artola
- Massachusetts General Hospital and the Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Justin S Sanchez
- Massachusetts General Hospital and Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Juliana Acosta-Uribe
- Grupo de Neurociencias de Antioquia de la Universidad de Antioquia, Medellin, Colombia
- Neuroscience Research Institute, Department of Molecular Cellular Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Matthew Lalli
- Neuroscience Research Institute, Department of Molecular Cellular Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Kenneth S Kosik
- Neuroscience Research Institute, Department of Molecular Cellular Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Matthew J Huentelman
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Rebecca A Reiman
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Ji Luo
- The Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Yinghua Chen
- The Banner Alzheimer's Institute, Phoenix, AZ, USA
| | | | - Yi Su
- The Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Gyungah R Jun
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Marcus Naymik
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Xiaowu Gai
- Center for Personalized Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Pathology, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Moiz Bootwalla
- Center for Personalized Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Jianling Ji
- Center for Personalized Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Pathology, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Lishuang Shen
- Center for Personalized Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - John B Miller
- Mass Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Leo A Kim
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Pierre N Tariot
- The Banner Alzheimer's Institute, Phoenix, AZ, USA
- University of Arizona, Tucson, AZ, USA
| | - Keith A Johnson
- Massachusetts General Hospital and Department of Neurology, Harvard Medical School, Boston, MA, USA
- Brigham and Women's Hospital and the Department of Neurology, Harvard Medical School, Boston, MA, USA
- Massachusetts General Hospital and Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Eric M Reiman
- The Banner Alzheimer's Institute, Phoenix, AZ, USA.
- University of Arizona, Tucson, AZ, USA.
- Arizona State University, Tempe, AZ, USA.
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ, USA.
| | - Yakeel T Quiroz
- Grupo de Neurociencias de Antioquia de la Universidad de Antioquia, Medellin, Colombia.
- Massachusetts General Hospital and Department of Neurology, Harvard Medical School, Boston, MA, USA.
- Massachusetts General Hospital and the Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
35
|
Hammond TR, Marsh SE, Stevens B. Immune Signaling in Neurodegeneration. Immunity 2019; 50:955-974. [PMID: 30995509 PMCID: PMC6822103 DOI: 10.1016/j.immuni.2019.03.016] [Citation(s) in RCA: 229] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/17/2019] [Accepted: 03/18/2019] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases of the central nervous system progressively rob patients of their memory, motor function, and ability to perform daily tasks. Advances in genetics and animal models are beginning to unearth an unexpected role of the immune system in disease onset and pathogenesis; however, the role of cytokines, growth factors, and other immune signaling pathways in disease pathogenesis is still being examined. Here we review recent genetic risk and genome-wide association studies and emerging mechanisms for three key immune pathways implicated in disease, the growth factor TGF-β, the complement cascade, and the extracellular receptor TREM2. These immune signaling pathways are important under both healthy and neurodegenerative conditions, and recent work has highlighted new functional aspects of their signaling. Finally, we assess future directions for immune-related research in neurodegeneration and potential avenues for immune-related therapies.
Collapse
Affiliation(s)
- Timothy R Hammond
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Samuel E Marsh
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Beth Stevens
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
36
|
Bostanciklioğlu M. The role of gut microbiota in pathogenesis of Alzheimer's disease. J Appl Microbiol 2019; 127:954-967. [PMID: 30920075 DOI: 10.1111/jam.14264] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/18/2019] [Accepted: 03/21/2019] [Indexed: 12/11/2022]
Abstract
This paper describes the effects of the gut microbiota on the pathogenesis of Alzheimer's pathology by evaluating the current original key findings and identifying gaps in the knowledge required for validation. The diversity of the gut microbiota declines in the elderly and in patients with Alzheimer's disease (AD). Restoring the diversity with probiotic treatment alleviates the psychiatric and histopathological findings. This presents a problem: How does gut microbiota interact with the pathogenesis of AD? The starting point of this comprehensive review is addressing the role of bacterial metabolites and neurotransmitters in the brain under various conditions, ranging from a healthy state to ageing and disease. In the light of current literature, we describe three different linkages between the present gut microbiome hypothesis and the other major theories for the pathogenesis of AD as follows: bacterial metabolites and amyloids can trigger central nervous system inflammation and cerebrovascular degeneration; impaired gut microbiome flora inhibits the autophagy-mediated protein clearance process; and gut microbiomes can change the neurotransmitter levels in the brain through the vagal afferent fibres.
Collapse
Affiliation(s)
- M Bostanciklioğlu
- Department of Physiology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| |
Collapse
|
37
|
Sirivichayakul S, Kanchanatawan B, Thika S, Carvalho AF, Maes M. A New Schizophrenia Model: Immune Activation is Associated with the Induction of Different Neurotoxic Products which Together Determine Memory Impairments and Schizophrenia Symptom Dimensions. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2019; 18:124-140. [DOI: 10.2174/1871527317666181119115532] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/02/2018] [Accepted: 11/13/2018] [Indexed: 12/19/2022]
Abstract
Objective: Recently, we reported that stable-phase schizophrenia is characterized by two interrelated
symptom dimensions: PHEMN (psychotic, hostility, excitation, mannerism and negative symptoms);
and DAPS (depressive, anxiety and physio-somatic symptoms) and that Major Neuro-Cognitive
psychosis (MNP) is the full-blown phenotype of schizophrenia (largely overlapping with deficit schizophrenia).
Herein we examined the effects of immune activation in association with tryptophan catabolite
(TRYCAT) patterning and memory disorders on PHEMN/DAPS dimensions and MNP.
Methods:
Serum levels of macrophage inflammatory protein-1 (MIP-1), soluble interleukin (IL)-1 receptor
antagonist (sIL-1RA), IL-10, eotaxin, IgA/IgM responses to TRYCATs, and Consortium to Establish
a Registry for Alzheimer’s disease (CERAD) tests were assessed in 40 controls and 80 schizophrenia
patients.
Results:
Schizophrenia and MNP were predicted by significantly increased levels of IL-10, eotaxin
and TRYCATs. A large part of variance in both PHEMN/DAPS symptom dimensions (42.8%) was
explained by cytokine levels and TRYCATs combined. The MIP+sIL-1RA+IL-10 composite score
and eotaxin explained each around on the basis of 19% of the variance in symptom dimensions, and
approximately 18% of memory deficits. Moreover, MIP+sIL-1RA+IL-10 was significantly associated
with elevations in picolinic acid, xanthurenic acid and 3-OH-kynurenine. Partial Least Squares path
modeling shows that highly significant effects of MIP+sIL-1RA+IL-10 on symptomatology are mediated
by the effects of noxious TRYCATs on memory deficits.
Conclusion:
Current findings indicate that in schizophrenia, immune activation may underpin activation
of indoleamine-2,3-dioxygenase and kynurenine monooxygenase, while impairments in episodic
and semantic memory may be caused by the neurotoxic effects of TRYCATs and eotaxin. The combined
effects of immune activation, eotaxin and memory defects determine to a large extent,
PHEMN/DAPS symptoms and the MNP phenotype. These findings indicate that schizophrenia phenomenology
is largely mediated by multiple neuro-immune pathways and that immune activation, increased
production of eotaxin and neurotoxic TRYCATs (picolinic acid, xanthurenic acid and 3-HOkynurenine)
are new drug targets in schizophrenia and MNP.
Collapse
Affiliation(s)
- Sunee Sirivichayakul
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Buranee Kanchanatawan
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Supaksorn Thika
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - André F. Carvalho
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW Over the last decade over 40 loci have been associated with risk of Alzheimer's disease (AD). However, most studies have either focused on identifying risk loci or performing unbiased screens without a focus on protective variation in AD. Here, we provide a review of known protective variants in AD and their putative mechanisms of action. Additionally, we recommend strategies for finding new protective variants. RECENT FINDINGS Recent Genome-Wide Association Studies have identified both common and rare protective variants associated with AD. These include variants in or near APP, APOE, PLCG2, MS4A, MAPT-KANSL1, RAB10, ABCA1, CCL11, SORL1, NOCT, SCL24A4-RIN3, CASS4, EPHA1, SPPL2A, and NFIC. SUMMARY There are very few protective variants with functional evidence and a derived allele with a frequency below 20%. Additional fine mapping and multi-omic studies are needed to further validate and characterize known variants as well as specialized genome-wide scans to identify novel variants.
Collapse
Affiliation(s)
- Shea J Andrews
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Equal first author
| | - Brian Fulton-Howard
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Equal first author
| | - Alison Goate
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
39
|
Association between plasma CCL11 (eotaxin-1) and cognitive status in older adults: Differences between rural and urban dwellers. Exp Gerontol 2018; 113:173-179. [PMID: 30308289 DOI: 10.1016/j.exger.2018.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/28/2018] [Accepted: 10/04/2018] [Indexed: 12/23/2022]
Abstract
The chemokine CCL11 has been implicated in age-related cognitive deterioration in mice, yet evidence on the relationship between CCL11 and cognitive function in humans is limited. This study explored associations between CCL11 and cognition in rural and urban community-dwelling older adults. Participants were 515 urban dwellers from the 3C-Bordeaux cohort and 318 rural dwellers from the AMI cohort. Plasma CCL11 was measured using an enzyme-linked immunoassay. Mini Mental State Examination (MMSE) test scores were used as the main measure of cognitive performance. Multivariate regression analysis was used to evaluate the cross-sectional association between CCL11 and cognitive performance. CCL11 was significantly higher in rural dwellers compared to city dwellers (median [IQR]: 145 [115-201] pg/mL vs. 103 [85-129] pg/mL; p < 0.001). After adjustment for confounders, CCL11 was found to be negatively associated with cognitive performance in rural dwellers but not in city dwellers. These results suggest that CCL11 may be an independent determinant of cognitive function in older rural dwellers and that the residential environment modifies this association.
Collapse
|
40
|
Sirivichayakul S, Kanchanatawan B, Thika S, Carvalho AF, Maes M. Eotaxin, an Endogenous Cognitive Deteriorating Chemokine (ECDC), Is a Major Contributor to Cognitive Decline in Normal People and to Executive, Memory, and Sustained Attention Deficits, Formal Thought Disorders, and Psychopathology in Schizophrenia Patients. Neurotox Res 2018; 35:122-138. [PMID: 30056534 DOI: 10.1007/s12640-018-9937-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 07/12/2018] [Accepted: 07/18/2018] [Indexed: 12/15/2022]
Abstract
Eotaxin is increased in neurodegenerative disorders and schizophrenia, and preclinical studies indicate that eotaxin may induce cognitive deficits. This study aims to examine whether peripheral levels of eotaxin impact cognitive functioning in healthy volunteers and formal thought disorder (FTD) and psychopathology in schizophrenia patients. Serum levels of eotaxin were assayed and cognitive tests were performed on a sample of 40 healthy participants and 80 schizophrenia patients. Among healthy participants, eotaxin levels were significantly associated with episodic/semantic memory, executive functions, Mini Mental State Examination, emotion recognition, and sustained attention. In addition, age-related effects on these cognitive measures were partly mediated by eotaxin. The super-variable "age-eotaxin" predicted a large part of the variance in cognitive functions among healthy participants, and hence, eotaxin may act as an "accelerated brain aging chemokine" (ABAC). In schizophrenia, eotaxin levels had a strong impact on formal thought disorders and psychopathology. In schizophrenia, increased eotaxin strongly impacts memory and sustained attention, which together to a large extent determine FTD. FTD together with memory deficits predicts around 92.5% of the variance in psychopathology. Moreover, the effects of eotaxin are partially mediated by executive functioning, while the effects of male sex on FTD and psychopathology are mediated by eotaxin. In healthy subjects, eotaxin strongly impacts executive functioning and multiple cognitive domains. In schizophrenia, peripheral levels of eotaxin strongly impact both negative symptoms and psychosis (hallucinations and delusions), and these eotaxin effects are mediated by impairments in frontal functioning, memory, sustained attention, and FTD. Eotaxin is an endogenous cognitive deteriorating chemokine (ECDC) and a novel therapeutic target for age-related cognitive decline and schizophrenia as well.
Collapse
Affiliation(s)
| | - Buranee Kanchanatawan
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Supaksorn Thika
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - André F Carvalho
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Michael Maes
- Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.
- IMPACT Strategic Research Center, Barwon Health, Deakin University, Geelong, VIC, Australia.
| |
Collapse
|
41
|
Blue EE, Yu CE, Thornton TA, Chapman NH, Kernfeld E, Jiang N, Shively KM, Buckingham KJ, Marvin CT, Bamshad MJ, Bird TD, Wijsman EM. Variants regulating ZBTB4 are associated with age-at-onset of Alzheimer's disease. GENES, BRAIN, AND BEHAVIOR 2018; 17:e12429. [PMID: 29045054 PMCID: PMC5902667 DOI: 10.1111/gbb.12429] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/11/2017] [Accepted: 10/12/2017] [Indexed: 01/01/2023]
Abstract
The identification of novel genetic modifiers of age-at-onset (AAO) of Alzheimer's disease (AD) could advance our understanding of AD and provide novel therapeutic targets. A previous genome scan for modifiers of AAO among families affected by early-onset AD caused by the PSEN2 N141I variant identified 2 loci with significant evidence for linkage: 1q23.3 and 17p13.2. Here, we describe the fine-mapping of these 2 linkage regions, and test for replication in 6 independent datasets. By fine-mapping these linkage signals in a single large family, we reduced the linkage regions to 11% their original size and nominated 54 candidate variants. Among the 11 variants associated with AAO of AD in a larger sample of Germans from Russia, the strongest evidence implicated promoter variants influencing NCSTN on 1q23.3 and ZBTB4 on 17p13.2. The association between ZBTB4 and AAO of AD was replicated by multiple variants in independent, trans-ethnic datasets. Our results show association between AAO of AD and both ZBTB4 and NCSTN. ZBTB4 is a transcriptional repressor that regulates the cell cycle, including the apoptotic response to amyloid beta, while NCSTN is part of the gamma secretase complex, known to influence amyloid beta production. These genes therefore suggest important roles for amyloid beta and cell cycle pathways in AAO of AD.
Collapse
Affiliation(s)
- Elizabeth E. Blue
- Division of Medical Genetics, University of Washington, Seattle, WA 98195, USA
| | - Chang-En Yu
- Division of Gerontology, University of Washington, Seattle, WA 98195, USA
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Timothy A. Thornton
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Nicola H. Chapman
- Division of Medical Genetics, University of Washington, Seattle, WA 98195, USA
| | - Eric Kernfeld
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Nan Jiang
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Kathryn M. Shively
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Kati J. Buckingham
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Colby T. Marvin
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Michael J. Bamshad
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Division of Genetic Medicine, Seattle Children’s Hospital, Seattle, WA 98105, USA
| | - Thomas D. Bird
- Division of Medical Genetics, University of Washington, Seattle, WA 98195, USA
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Neurology, University of Washington, Seattle, WA 98195, USA
| | - Ellen M. Wijsman
- Division of Medical Genetics, University of Washington, Seattle, WA 98195, USA
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
42
|
Zhu X, Yu H, Xiao Q, Ke J, Li H, Chen Z, Ding H, Leng S, Huang Y, Zhan J, Lei J, Fan W, Luo H. Genetic variations in chromodomain helicase DNA-binding protein 5, gene-environment interactions and risk of sporadic Alzheimer's disease in Chinese population. Oncotarget 2018; 9:24872-24881. [PMID: 29861839 PMCID: PMC5982770 DOI: 10.18632/oncotarget.23791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 12/05/2017] [Indexed: 11/25/2022] Open
Abstract
CHD5 is an essential factor for neuronal differentiation and neurodegenerative diseases. Here, the targeted next generation sequencing and TaqMan genotyping technologies were carried out for CHD5 gene in a two-staged case-control study in Chinese population. The genetic statistics and gene-environment interactions were analyzed to find certain risk factors of Alzheimer's disease. We found intronic rs11121295 was associated with the risk of Alzheimer's disease at both stages including combined cohorts. This risk effect presented consistently significant associations with the alcoholic subgroups at both all stages in the stratified analysis. The gene-environment interactions further supported the above findings. Our study highlighted the potential role of CHD5 variants in conferring susceptibility to sporadic Alzheimer's disease, especially modified its risk by alcoholic intake.
Collapse
Affiliation(s)
- Xiao Zhu
- Key Laboratory of Medical Molecular Diagnosis, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, China.,Institute of Bioinformatics, University of Georgia, Athens, GA, USA
| | - Haibing Yu
- Key Laboratory of Medical Molecular Diagnosis, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, China
| | - Qin Xiao
- Department of Blood Transfusion, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jianhao Ke
- Tropical Crops Department, Guangdong AIB Polytechnic, Guangzhou, China
| | - Hongmei Li
- Key Laboratory of Medical Molecular Diagnosis, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, China
| | - Zhihong Chen
- Key Laboratory of Medical Molecular Diagnosis, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, China
| | - Hongrong Ding
- Key Laboratory of Medical Molecular Diagnosis, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, China
| | - Shuilong Leng
- Department of Human Anatomy, Guangzhou Medical University, Guangzhou, China
| | - Yongmei Huang
- Institute of Marine Medicine Research, Guangdong Medical University, Zhanjiang, China
| | - Jingting Zhan
- Institute of Marine Medicine Research, Guangdong Medical University, Zhanjiang, China
| | - Jinli Lei
- Institute of Marine Medicine Research, Guangdong Medical University, Zhanjiang, China
| | - Wenguo Fan
- Department of Anatomy and Physiology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Hui Luo
- Key Laboratory of Medical Molecular Diagnosis, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, China.,Institute of Marine Medicine Research, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
43
|
Kang WS, Kim YJ, Park HJ, Kim SK, Paik JW, Kim JW. Association of CCL11 promoter polymorphisms with schizophrenia in a Korean population. Gene 2018; 656:80-85. [PMID: 29477870 DOI: 10.1016/j.gene.2018.02.053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 02/12/2018] [Accepted: 02/21/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Immunological alterations and dysregulation of the inflammatory response have been suggested to play a crucial role in schizophrenia pathophysiology. Growing evidence supports the involvement of chemokines in brain development, thus many chemokines have been studied in relation with schizophrenia. The C-C motif chemokine ligand 11 (CCL11) has been shown to be related with synaptic plasticity and neurogenesis. Moreover, altered levels of CCL11 have been observed in schizophrenia patients. Therefore, we examined whether single nucleotide polymorphisms (SNPs) of the CCL11 in the promoter region contribute to susceptibility to schizophrenia. METHODS Four promoter SNPs [rs17809012 (-384T>C), rs16969415 (-426C>T), rs17735961 (-488C>A), and rs4795896 (576G>A)] were genotyped in 254 schizophrenia patients and 405 control subjects using Fluidigm SNPtype assays. RESULTS The genotype frequency of CCL11 rs4795896 (-576G>A) showed significant association with schizophrenia in a recessive model (AA vs. GG/AG, p < 0.0001) and in a log-additive model (AG vs. AA vs. GG, p < 0.0001). The allele frequency of rs4795896 also showed a significant association with schizophrenia (p < 0.0001). Furthermore, haplotype analysis revealed that GCT, ACT, and GCC haplotypes containing rs4795896, rs17735961 and rs17809012 were significantly associated with schizophrenia (p = 0.0044, p < 0.0001, and p < 0.0001, respectively). CONCLUSION Our results suggest that CCL11 promotor polymorphism is associated with increased risk for the development of schizophrenia in a Korean population.
Collapse
Affiliation(s)
- Won Sub Kang
- Department of Psychiatry, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Young Jong Kim
- Department of Psychiatry, Myongii Hospital, Goyang-si, Gyeonggi-do 10475, Republic of Korea
| | - Hae Jeong Park
- Kohwang Medical Research Institute, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Su Kang Kim
- Kohwang Medical Research Institute, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jong-Woo Paik
- Department of Psychiatry, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jong Woo Kim
- Department of Psychiatry, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
44
|
Kuo HW, Liu TH, Tsou HH, Hsu YT, Wang SC, Fang CP, Liu CC, Chen ACH, Liu YL. Inflammatory chemokine eotaxin-1 is correlated with age in heroin dependent patients under methadone maintenance therapy. Drug Alcohol Depend 2018; 183:19-24. [PMID: 29222992 DOI: 10.1016/j.drugalcdep.2017.10.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/03/2017] [Accepted: 10/07/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Degeneration of central neurons and fibers has been observed in postmortem brains of heroin dependent patients. However, there are no biomarkers to predict the severity of neurodegeneration related to heroin dependence. A correlation has been reported between inflammatory C-C motif chemokine ligand 11 (CCL11, or eotaxin-1) and neurodegeneration in Alzheimer's disease. METHODS Three-hundred-forty-four heroin dependent, Taiwanese patients under methadone maintenance treatment (MMT) were included with clinical assessment and genomics information. Eighty-seven normal control subjects were also recruited for comparison. RESULTS Using receiver operating characteristics curve analyses, CCL11 showed the strongest sensitivity and specificity in correlation with age by a cut-off at 45 years (AUC = 0.69, P < 0.0001) in MMT patients, but not normal controls. Patients 45 years of age or older had significantly higher plasma levels of CCL11, fibroblast growth factor 2 (FGF-2), nicotine metabolite cotinine, and a longer duration of addiction. Plasma level of CCL11 was correlated with that of FGF-2 (partial r2 = 0.24, P < 0.0001). Carriers with the mutant allele of rs1129844, a functional single nucleotide polymorphism (Ala23Thr) in the CCL11 gene, showed a higher plasma level of Aß42, ratio of Aß42/Aß40, and insomnia side effect symptom score than the GG genotype carriers among MMT responders with morphine-negative urine results. CONCLUSIONS The results suggest possible novel mechanisms mediated through CCL11 involving neurotoxicity in heroin dependent patients.
Collapse
Affiliation(s)
- Hsiang-Wei Kuo
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Tung-Hsia Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Hsiao-Hui Tsou
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Miaoli County, Taiwan; Graduate Institute of Biostatistics, College of Public Health, China Medical University, Taichung, Taiwan
| | - Ya-Ting Hsu
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Miaoli County, Taiwan
| | - Sheng-Chang Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Chiu-Ping Fang
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Chia-Chen Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Andrew C H Chen
- Department of Psychiatry, The Zucker Hillside Hospital, Northwell Health System, Glen Oaks, NY, USA; The Feinstein Institute for Medical Research, Hofstra Northwell School of Medicine at Hofstra University, Manhasset, NY, USA
| | - Yu-Li Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Taiwan; Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.
| |
Collapse
|
45
|
Minter MR, Hinterleitner R, Meisel M, Zhang C, Leone V, Zhang X, Oyler-Castrillo P, Zhang X, Musch MW, Shen X, Jabri B, Chang EB, Tanzi RE, Sisodia SS. Antibiotic-induced perturbations in microbial diversity during post-natal development alters amyloid pathology in an aged APP SWE/PS1 ΔE9 murine model of Alzheimer's disease. Sci Rep 2017; 7:10411. [PMID: 28874832 PMCID: PMC5585265 DOI: 10.1038/s41598-017-11047-w] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 08/18/2017] [Indexed: 02/06/2023] Open
Abstract
Recent evidence suggests the commensal microbiome regulates host immunity and influences brain function; findings that have ramifications for neurodegenerative diseases. In the context of Alzheimer’s disease (AD), we previously reported that perturbations in microbial diversity induced by life-long combinatorial antibiotic (ABX) selection pressure in the APPSWE/PS1ΔE9 mouse model of amyloidosis is commensurate with reductions in amyloid-β (Aβ) plaque pathology and plaque-localised gliosis. Considering microbiota-host interactions, specifically during early post-natal development, are critical for immune- and neuro-development we now examine the impact of microbial community perturbations induced by acute ABX exposure exclusively during this period in APPSWE/PS1ΔE9 mice. We show that early post-natal (P) ABX treatment (P14-P21) results in long-term alterations of gut microbial genera (predominantly Lachnospiraceae and S24-7) and reduction in brain Aβ deposition in aged APPSWE/PS1ΔE9 mice. These mice exhibit elevated levels of blood- and brain-resident Foxp3+ T-regulatory cells and display an alteration in the inflammatory milieu of the serum and cerebrospinal fluid. Finally, we confirm that plaque-localised microglia and astrocytes are reduced in ABX-exposed mice. These findings suggest that ABX-induced microbial diversity perturbations during post-natal stages of development coincide with altered host immunity mechanisms and amyloidosis in a murine model of AD.
Collapse
Affiliation(s)
- Myles R Minter
- Department of Neurobiology, The University of Chicago, Chicago, IL, 60637, USA.,The Microbiome Center, The University of Chicago, Chicago, IL, 60637, USA
| | - Reinhard Hinterleitner
- Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA.,Committee on Immunology, The University of Chicago, Chicago, IL, 60637, USA
| | - Marlies Meisel
- Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA.,Committee on Immunology, The University of Chicago, Chicago, IL, 60637, USA
| | - Can Zhang
- Department of Neurology, Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Diseases, Massachusetts General Hospital, Charlestown, MA, 02114, USA
| | - Vanessa Leone
- The Microbiome Center, The University of Chicago, Chicago, IL, 60637, USA.,Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Xiaoqiong Zhang
- Department of Neurobiology, The University of Chicago, Chicago, IL, 60637, USA
| | | | - Xulun Zhang
- Department of Neurobiology, The University of Chicago, Chicago, IL, 60637, USA
| | - Mark W Musch
- Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Xunuo Shen
- Department of Neurology, Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Diseases, Massachusetts General Hospital, Charlestown, MA, 02114, USA
| | - Bana Jabri
- Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA.,Committee on Immunology, The University of Chicago, Chicago, IL, 60637, USA
| | - Eugene B Chang
- The Microbiome Center, The University of Chicago, Chicago, IL, 60637, USA.,Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Rudolph E Tanzi
- Department of Neurology, Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Diseases, Massachusetts General Hospital, Charlestown, MA, 02114, USA
| | - Sangram S Sisodia
- Department of Neurobiology, The University of Chicago, Chicago, IL, 60637, USA. .,The Microbiome Center, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
46
|
Gene expression patterns associated with neurological disease in human HIV infection. PLoS One 2017; 12:e0175316. [PMID: 28445538 PMCID: PMC5405951 DOI: 10.1371/journal.pone.0175316] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 03/23/2017] [Indexed: 12/01/2022] Open
Abstract
The pathogenesis and nosology of HIV-associated neurological disease (HAND) remain incompletely understood. Here, to provide new insight into the molecular events leading to neurocognitive impairments (NCI) in HIV infection, we analyzed pathway dysregulations in gene expression profiles of HIV-infected patients with or without NCI and HIV encephalitis (HIVE) and control subjects. The Gene Set Enrichment Analysis (GSEA) algorithm was used for pathway analyses in conjunction with the Molecular Signatures Database collection of canonical pathways (MSigDb). We analyzed pathway dysregulations in gene expression profiles of patients from the National NeuroAIDS Tissue Consortium (NNTC), which consists of samples from 3 different brain regions, including white matter, basal ganglia and frontal cortex of HIV-infected and control patients. While HIVE is characterized by widespread, uncontrolled inflammation and tissue damage, substantial gene expression evidence of induction of interferon (IFN), cytokines and tissue injury is apparent in all brain regions studied, even in the absence of NCI. Various degrees of white matter changes were present in all HIV-infected subjects and were the primary manifestation in patients with NCI in the absence of HIVE. In particular, NCI in patients without HIVE in the NNTC sample is associated with white matter expression of chemokines, cytokines and β-defensins, without significant activation of IFN. Altogether, the results identified distinct pathways differentially regulated over the course of neurological disease in HIV infection and provide a new perspective on the dynamics of pathogenic processes in the course of HIV neurological disease in humans. These results also demonstrate the power of the systems biology analyses and indicate that the establishment of larger human gene expression profile datasets will have the potential to provide novel mechanistic insight into the pathogenesis of neurological disease in HIV infection and identify better therapeutic targets for NCI.
Collapse
|
47
|
Bolós M, Perea JR, Avila J. Alzheimer's disease as an inflammatory disease. Biomol Concepts 2017; 8:37-43. [PMID: 28231054 DOI: 10.1515/bmc-2016-0029] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 01/10/2017] [Indexed: 02/08/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition characterized by the formation of amyloid-β plaques, aggregated and hyperphosphorylated tau protein, activated microglia and neuronal cell death, ultimately leading to progressive dementia. In this short review, we focus on neuroinflammation in AD. Specifically, we describe the participation of microglia, as well as other factors that may contribute to inflammation, in neurodegeneration.
Collapse
Affiliation(s)
- Marta Bolós
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Valderrebollo 5, E-28041-Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa" CSIC-UAM, Nicolás Cabrera 1, E-28049-Madrid, Spain
| | - Juan Ramón Perea
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Valderrebollo 5, E-28041-Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa" CSIC-UAM, Nicolás Cabrera 1, E-28049-Madrid, Spain
| | - Jesús Avila
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Valderrebollo 5, E-28041-Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa" CSIC-UAM, Nicolás Cabrera 1, E-28049-Madrid, Spain
| |
Collapse
|
48
|
Natelson Love M, Clark DG, Cochran JN, Den Beste KA, Geldmacher DS, Benzinger TL, Gordon BA, Morris JC, Bateman RJ, Roberson ED. Clinical, imaging, pathological, and biochemical characterization of a novel presenilin 1 mutation (N135Y) causing Alzheimer's disease. Neurobiol Aging 2017; 49:216.e7-216.e13. [PMID: 27793474 PMCID: PMC5154842 DOI: 10.1016/j.neurobiolaging.2016.09.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 08/21/2016] [Accepted: 09/24/2016] [Indexed: 01/19/2023]
Abstract
We present 2 cases of early-onset Alzheimer's disease due to a novel N135Y mutation in PSEN1. The proband presented with memory and other cognitive symptoms at age 32. Detailed clinical characterization revealed initial deficits in memory with associated dysarthria, progressing to involve executive dysfunction, spastic gait, and episodic confusion with polyspike discharges on long-term electroencephalography. Amyloid- and FDG-PET scans showed typical results of Alzheimer's disease. By history, the proband's father had developed cognitive symptoms at age 42 and died at age 48. Neuropathological evaluation confirmed Alzheimer's disease, with moderate to severe amyloid angiopathy. Skeletal muscle showed type 2 fiber-predominant atrophy with pale central clearing. Genetic testing of the proband revealed an N135Y missense mutation in PSEN1. This mutation was predicted to be pathogenic by in silico analysis. Biochemical analysis confirmed that the mutation caused an increased Aβ42/Aβ40 ratio, consistent with other PSEN1 mutations and with a loss of presenilin function.
Collapse
Affiliation(s)
- Marissa Natelson Love
- Alzheimer's Disease Center, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Neurology, Birmingham VA Medical Center, Birmingham, AL, USA
| | - David G Clark
- Alzheimer's Disease Center, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Neurology, Birmingham VA Medical Center, Birmingham, AL, USA; Department of Neurology, Ralph H. Johnson VA Medical Center, Medical University of South Carolina, Charleston, SC, USA.
| | - J Nicholas Cochran
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kyle A Den Beste
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David S Geldmacher
- Alzheimer's Disease Center, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA; McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tammie L Benzinger
- Dominantly Inherited Alzheimer's Network, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Dominantly Inherited Alzheimer's Network, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Dominantly Inherited Alzheimer's Network, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J Bateman
- Dominantly Inherited Alzheimer's Network, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Erik D Roberson
- Alzheimer's Disease Center, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA; McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
49
|
Wyss-Coray T. Ageing, neurodegeneration and brain rejuvenation. Nature 2016; 539:180-186. [PMID: 27830812 PMCID: PMC5172605 DOI: 10.1038/nature20411] [Citation(s) in RCA: 741] [Impact Index Per Article: 82.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/02/2016] [Indexed: 02/08/2023]
Abstract
Although systemic diseases take the biggest toll on human health and well-being, increasingly, a failing brain is the arbiter of a death preceded by a gradual loss of the essence of being. Ageing, which is fundamental to neurodegeneration and dementia, affects every organ in the body and seems to be encoded partly in a blood-based signature. Indeed, factors in the circulation have been shown to modulate ageing and to rejuvenate numerous organs, including the brain. The discovery of such factors, the identification of their origins and a deeper understanding of their functions is ushering in a new era in ageing and dementia research.
Collapse
Affiliation(s)
- Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Palo Alto, California 94304, USA
- Center for Tissue Regeneration, Repair and Restoration, VA Palo Alto Health Care System, Palo Alto, California 94304, USA
| |
Collapse
|
50
|
Grewal R, Haghighi M, Huang S, Smith AG, Cao C, Lin X, Lee DC, Teten N, Hill AM, Selenica MLB. Identifying biomarkers of dementia prevalent among amnestic mild cognitively impaired ethnic female patients. ALZHEIMERS RESEARCH & THERAPY 2016; 8:43. [PMID: 27756387 PMCID: PMC5067885 DOI: 10.1186/s13195-016-0211-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 09/13/2016] [Indexed: 12/22/2022]
Abstract
Background There is a need to investigate biomarkers that are indicative of the progression of dementia in ethnic patient populations. The disparity of information in these populations has been the focus of many clinical and academic centers, including ours, to contribute to a higher success rate in clinical trials. In this study, we have investigated plasma biomarkers in amnestic mild cognitively impaired (aMCI) female patient cohorts in the context of ethnicity and cognitive status. Method A panel of 12 biomarkers involved in the progression of brain pathology, inflammation, and cardiovascular disorders were investigated in female cohorts of African American, Hispanic, and White aMCI patients. Both biochemical and algorithmic analyses were applied to correlate biomarker levels measured during the early stages of the disease for each ethnicity. Results We report elevated plasma Aβ40, Aβ42, YKL-40, and cystatin C levels in the Hispanic cohort at early aMCI status. In addition, elevated plasma Aβ40 levels were associated with the aMCI status in both White and African American patient cohorts by the decision tree algorithm. Eotaxin-1 levels, as determined by the decision tree algorithm and biochemically measured total tau levels, were associated with the aMCI status in the African American cohort. Conclusions Overall, our data displayed novel differences in the plasma biomarkers of the aMCI female cohorts where the plasma levels of several biomarkers distinguished between each ethnicity at an early aMCI stage. Identification of these plasma biomarkers encourages new areas of investigation among aMCI ethnic populations, including larger patient cohorts and longitudinal study designs. Electronic supplementary material The online version of this article (doi:10.1186/s13195-016-0211-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rinko Grewal
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Mona Haghighi
- Department of Industrial and Systems Engineering, University of Washington, 3900 Northeast Stevens Way, Seattle, WA, 98195, USA
| | - Shuai Huang
- Department of Industrial and Systems Engineering, University of Washington, 3900 Northeast Stevens Way, Seattle, WA, 98195, USA.,School of Aging Studies, University of South Florida, 4202 E Fowler Ave, Tampa, FL, 33620, USA
| | - Amanda G Smith
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Psychiatry and Behavioral Medicine, College of Medicine, University of South Florida, 3515 E Fletcher Ave, Tampa, FL, 33613, USA
| | - Chuanhai Cao
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Xiaoyang Lin
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Daniel C Lee
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Nancy Teten
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Angela M Hill
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Pharmacotherapeutics and Clinical Research, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Maj-Linda B Selenica
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA. .,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|