1
|
Galkin F, Pulous FE, Fu Y, Zhang M, Pun FW, Ren F, Zhavoronkov A. Roles of hypoxia-inducible factor-prolyl hydroxylases in aging and disease. Ageing Res Rev 2024; 102:102551. [PMID: 39447706 DOI: 10.1016/j.arr.2024.102551] [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: 05/08/2024] [Revised: 10/07/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
The prolyl hydroxylase domain-containing (PHD or EGL9-homologs) enzyme family is mainly known for its role in the cellular response to hypoxia. HIF-PH inhibitors can stabilize hypoxia-inducible factors (HIFs), activating transcriptional programs that promote processes such as angiogenesis and erythropoiesis to adapt to changes in oxygen levels. HIF-PH inhibitors have been clinically approved for treating several types of anaemia. While most discussions of the HIF-PH signalling axis focus on hypoxia, there is a growing recognition of its importance under normoxic conditions. Recent advances in PHD biology have highlighted the potential of targeting this pathway therapeutically for a range of aging-related diseases. In this article, we review these recent discoveries, situate them within the broader context of aging and disease, and explore current therapeutic strategies that target PHD enzymes for these indications.
Collapse
Affiliation(s)
- Fedor Galkin
- Insilico Medicine AI Ltd., Level 6, Unit 08, Block A, IRENA HQ Building, Masdar City, Abu Dhabi, UAE
| | - Fadi E Pulous
- Insilico Medicine US Inc., 1000 Massachusetts Avenue, Suite 126, Cambridge, MA 02138, United States
| | - Yanyun Fu
- Insilico Medicine Shanghai Ltd., Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong, Shanghai 201203, China
| | - Man Zhang
- Insilico Medicine Shanghai Ltd., Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong, Shanghai 201203, China
| | - Frank W Pun
- Insilico Medicine Hong Kong Ltd., Unit 310, 3/F, Building 8W, Hong Kong Science and Technology Park, Hong Kong SAR
| | - Feng Ren
- Insilico Medicine AI Ltd., Level 6, Unit 08, Block A, IRENA HQ Building, Masdar City, Abu Dhabi, UAE; Insilico Medicine Shanghai Ltd., Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong, Shanghai 201203, China; Insilico Medicine Hong Kong Ltd., Unit 310, 3/F, Building 8W, Hong Kong Science and Technology Park, Hong Kong SAR
| | - Alex Zhavoronkov
- Insilico Medicine AI Ltd., Level 6, Unit 08, Block A, IRENA HQ Building, Masdar City, Abu Dhabi, UAE; Insilico Medicine US Inc., 1000 Massachusetts Avenue, Suite 126, Cambridge, MA 02138, United States; Insilico Medicine Hong Kong Ltd., Unit 310, 3/F, Building 8W, Hong Kong Science and Technology Park, Hong Kong SAR; Insilico Medicine Canada Inc., 1250 René-Lévesque Ouest, Suite 3710, Montréal, Québec H3B 4W8, Canada; Buck Institute for Research on Aging, Novato, CA, United States.
| |
Collapse
|
2
|
Fanlo-Ucar H, Picón-Pagès P, Herrera-Fernández V, ILL-Raga G, Muñoz FJ. The Dual Role of Amyloid Beta-Peptide in Oxidative Stress and Inflammation: Unveiling Their Connections in Alzheimer's Disease Etiopathology. Antioxidants (Basel) 2024; 13:1208. [PMID: 39456461 PMCID: PMC11505517 DOI: 10.3390/antiox13101208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, and it is currently the seventh leading cause of death worldwide. It is characterized by the extracellular aggregation of the amyloid β-peptide (Aβ) into oligomers and fibrils that cause synaptotoxicity and neuronal death. Aβ exhibits a dual role in promoting oxidative stress and inflammation. This review aims to unravel the intricate connection between these processes and their contribution to AD progression. The review delves into oxidative stress in AD, focusing on the involvement of metals, mitochondrial dysfunction, and biomolecule oxidation. The distinct yet overlapping concept of nitro-oxidative stress is also discussed, detailing the roles of nitric oxide, mitochondrial perturbations, and their cumulative impact on Aβ production and neurotoxicity. Inflammation is examined through astroglia and microglia function, elucidating their response to Aβ and their contribution to oxidative stress within the AD brain. The blood-brain barrier and oligodendrocytes are also considered in the context of AD pathophysiology. We also review current diagnostic methodologies and emerging therapeutic strategies aimed at mitigating oxidative stress and inflammation, thereby offering potential treatments for halting or slowing AD progression. This comprehensive synthesis underscores the pivotal role of Aβ in bridging oxidative stress and inflammation, advancing our understanding of AD and informing future research and treatment paradigms.
Collapse
Affiliation(s)
- Hugo Fanlo-Ucar
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| | - Pol Picón-Pagès
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
- Laboratory of Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08028 Barcelona, Spain
| | - Víctor Herrera-Fernández
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| | - Gerard ILL-Raga
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| | - Francisco J. Muñoz
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| |
Collapse
|
3
|
Diniz DG, Bento-Torres J, da Costa VO, Carvalho JPR, Tomás AM, Galdino de Oliveira TC, Soares FC, de Macedo LDED, Jardim NYV, Bento-Torres NVO, Anthony DC, Brites D, Picanço Diniz CW. The Hidden Dangers of Sedentary Living: Insights into Molecular, Cellular, and Systemic Mechanisms. Int J Mol Sci 2024; 25:10757. [PMID: 39409085 PMCID: PMC11476792 DOI: 10.3390/ijms251910757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
With the aging of the global population, neurodegenerative diseases are emerging as a major public health issue. The adoption of a less sedentary lifestyle has been shown to have a beneficial effect on cognitive decline, but the molecular mechanisms responsible are less clear. Here we provide a detailed analysis of the complex molecular, cellular, and systemic mechanisms underlying age-related cognitive decline and how lifestyle choices influence these processes. A review of the evidence from animal models, human studies, and postmortem analyses emphasizes the importance of integrating physical exercise with cognitive, multisensory, and motor stimulation as part of a multifaceted approach to mitigating cognitive decline. We highlight the potential of these non-pharmacological interventions to address key aging hallmarks, such as genomic instability, telomere attrition, and neuroinflammation, and underscore the need for comprehensive and personalized strategies to promote cognitive resilience and healthy aging.
Collapse
Affiliation(s)
- Daniel Guerreiro Diniz
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Seção de Hepatologia, Belém 66.093-020, Pará, Brazil;
- Núcleo de Pesquisas em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil;
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
| | - João Bento-Torres
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
- Programa de Pós-Graduação em Ciências do Movimento Humano, Universidade Federal do Pará, Belém 66.050-160, Pará, Brazil
| | - Victor Oliveira da Costa
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
| | - Josilayne Patricia Ramos Carvalho
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
- Programa de Pós-Graduação em Ciências do Movimento Humano, Universidade Federal do Pará, Belém 66.050-160, Pará, Brazil
| | - Alessandra Mendonça Tomás
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
- Campus Samabaia, Universidade Federal de Goiás (EBTT), CEPAE, Goiânia 74.001-970, Goiás, Brazil
| | - Thaís Cristina Galdino de Oliveira
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
- Faculdade de Ceilândia, Ceilândia, Universidade de Brasília, Brasília 72.220-900, Brazil
| | - Fernanda Cabral Soares
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
| | - Liliane Dias e Dias de Macedo
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
- Campus Tucurui, Universidade do Estado do Pará, Tucurui 68.455-210, Pará, Brazil
| | - Naina Yuki Vieira Jardim
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
- Campus Tucurui, Universidade do Estado do Pará, Tucurui 68.455-210, Pará, Brazil
- Programa de Pós-Graduação em Neurociências e Biologia Celular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66.075-110, Pará, Brazil
| | - Natáli Valim Oliver Bento-Torres
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
- Programa de Pós-Graduação em Ciências do Movimento Humano, Universidade Federal do Pará, Belém 66.050-160, Pará, Brazil
| | - Daniel Clive Anthony
- Laboratory of Experimental Neuropathology, Department of Pharmacology, University of Oxford, Oxford OX1 2JD, UK;
| | - Dora Brites
- Faculty of Pharmacy, Department of Pharmaceutical Sciences and Medicines, Universidade de Lisboa, 1649-003 Lisbon, Portugal;
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Cristovam Wanderley Picanço Diniz
- Núcleo de Pesquisas em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil;
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
- Programa de Pós-Graduação em Neurociências e Biologia Celular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66.075-110, Pará, Brazil
| |
Collapse
|
4
|
Nasr El-Din WA, Abdel Fattah IO. L-arginine mitigates choroid plexus changes in Alzheimer's disease rat model via oxidative/inflammatory burden and behavioral modulation. Tissue Cell 2024; 91:102572. [PMID: 39326233 DOI: 10.1016/j.tice.2024.102572] [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: 08/14/2024] [Revised: 09/07/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Aging is a risk factor for Alzheimer's disease (AD), leading to choroid plexus (CP) alterations. This study aimed to explore the possible therapeutic mechanisms of ARG on AD-induced CP changes. Sprague-Dawley rats were divided into 6 groups (n = 7 per group): adult, adult+ARG, aged, aged+ARG, aged+AD, and aged+AD+ARG groups. Evaluations were for Y-maze test, serum levels of oxidative/inflammatory markers, and serum and cerebrospinal fluid (CSF) markers of AD, histopathology, immunohistochemistry, and histomorphometry. The aged+AD group demonstrated a significant decline in maze test parameters, total antioxidant capacity (TAC), brain-derived neurotrophic factor (BDNF) levels, and vascular endothelial growth factor (VEGF) immunoexpression, while tumour necrosis factor-α (TNF-α), interleukin-1 beta (IL-1β), beta-amyloid (Aβ) levels and amyloid protein precursor (APP), and heat shock protein90 (HSP90) immunoexpressions were significantly increased. Sections of this group showed flat epitheliocytes, congested capillaries, connective tissue expansion, and degenerated endothelium. These parameters were modulated by ARG administration, via increased levels of TAC (1.37 vs 2.17 mmol/L), (p = 0.018) BDNF (serum: 48.50 vs 78.41; CSF: 4.07 vs 7.11 pg/ml) (p< 0.001), and VEGF (0.07 vs 0.26 OD) (p< 0.001), in addition to decreased levels of TNF-α (86.63 vs 41.39 pg/ml) (p< 0.001), IL-1β (96.04 vs 39.57 pg/ml) (p< 0.001), Aβ (serum: 67.40 vs 47.30; CSF: 189.26 vs 169.84 pg/ml) (p< 0.001), and HSP90 (0.54 vs 0.13 OD) (p< 0.001). In conclusion, ARG ameliorates the AD-associated CP changes, including histopathological, oxidative/inflammatory, and AD markers, and VEGF and HSP90 immunohistochemical alterations. Dietary ARG consumption is recommended to avoid AD progression in the elderly.
Collapse
Affiliation(s)
- Wael Amin Nasr El-Din
- Department of Anatomy, College of Medicine and Health Sciences, Arabian Gulf University, Manama, Bahrain; Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Islam Omar Abdel Fattah
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| |
Collapse
|
5
|
Cerilli E, Dall'O GM, Chelini G, Catena B, Weinberger B, Bozzi Y, Pangrazzi L. Immune system dysfunction and inflammation in aging Shank3b mutant mice, a model of autism spectrum disorder. Front Immunol 2024; 15:1447385. [PMID: 39308859 PMCID: PMC11412883 DOI: 10.3389/fimmu.2024.1447385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/12/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction Autism spectrum disorder (ASD) is a heterogeneous group of neurodevelopmental Q8 conditions characterized by deficits in social interaction/communication and restrictive/repetitive behaviors. Recent studies highlight the role of immune system dysfunction and inflammation in ASD pathophysiology. Indeed, elevated levels of pro-inflammatory cytokines were described in the brain and peripheral blood of ASD individuals. Despite this, how this pro-inflammatory profile evolves with aging and whether it may be associated with behavioral deficits is unknown. In this work, we explored the impact of aging on motor behavior and inflammation using Shank3b mutant mice, a model for syndromic ASD. Methods Using RT-qPCR and flow cytometry, we examined the expression of key pro-inflammatory molecules in the cerebellum, bone marrow, spleen, and peripheral blood, comparing adult and old Shank3b +/+, Shank3b +/-, and Shank3b -/- mice. Results and discussion Our findings revealed genotype- and age-related differences in inflammation and motor behavior, with Shank3b-/- mice exhibiting accelerated aging and motor impairments. Correlations between pro-inflammatory molecules and behavioral deficits suggest that a link may be present between systemic inflammation and ASD-related behaviors, underscoring the potential role of age-related inflammation ("inflammaging") in exacerbating ASD symptoms.
Collapse
Affiliation(s)
- Enrica Cerilli
- CIMeC - Center for Mind/Brain Sciences, University of Trento, Rovereto, Trento, Italy
| | | | - Gabriele Chelini
- CIMeC - Center for Mind/Brain Sciences, University of Trento, Rovereto, Trento, Italy
- Department of Biomedical Sciences, CNR Neuroscience Institute, Pisa, Italy
| | - Benedetta Catena
- CIMeC - Center for Mind/Brain Sciences, University of Trento, Rovereto, Trento, Italy
| | - Birgit Weinberger
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Yuri Bozzi
- CIMeC - Center for Mind/Brain Sciences, University of Trento, Rovereto, Trento, Italy
- Department of Biomedical Sciences, CNR Neuroscience Institute, Pisa, Italy
| | - Luca Pangrazzi
- CIMeC - Center for Mind/Brain Sciences, University of Trento, Rovereto, Trento, Italy
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
6
|
Frank G, Gualtieri P, Cianci R, Caldarelli M, Palma R, De Santis GL, Porfilio C, Nicoletti F, Bigioni G, Di Renzo L. Body Composition and Alzheimer's Disease: A Holistic Review. Int J Mol Sci 2024; 25:9573. [PMID: 39273520 PMCID: PMC11395597 DOI: 10.3390/ijms25179573] [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: 07/25/2024] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Alzheimer's disease (AD) represents a significant global health challenge and affects approximately 50 million people worldwide. This overview of published reviews provides a comprehensive understanding of the intricate correlations between AD and body composition, focusing particularly on obesity. We used a systematic approach to collect and analyze relevant reviews on the topic of obesity and Alzheimer's disease. A comprehensive search of electronic databases, including PubMed, MEDLINE, and Google Scholar, was conducted. We searched keywords such as "Alzheimer's disease", "body composition", "lean mass", "bone mass", and "fat mass". We considered only reviews written within the past 5 years and in English. Fifty-six relevant reviews were identified that shed light on the multiple connections between AD and body composition. The review involves several aspects, including the impact of lean mass, bone mass, and endocrinological factors related to obesity, as well as inflammation, neuroinflammation, and molecular/genetic factors. The findings highlight the complex interplay of these elements in the development of AD, underscoring the need for holistic approaches to reduce the risk of AD and to explore innovative strategies for diagnosis, prevention, and treatment.
Collapse
Affiliation(s)
- Giulia Frank
- PhD School of Applied Medical-Surgical Sciences, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
- School of Specialization in Food Science, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Paola Gualtieri
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Rossella Cianci
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Mario Caldarelli
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Roselisa Palma
- PhD School of Applied Medical-Surgical Sciences, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
- School of Specialization in Food Science, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Gemma Lou De Santis
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Chiara Porfilio
- School of Specialization in Food Science, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Francesco Nicoletti
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Giulia Bigioni
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Laura Di Renzo
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|
7
|
Dou L, Peng Y, Zhang B, Yang H, Zheng K. Immune Remodeling during Aging and the Clinical Significance of Immunonutrition in Healthy Aging. Aging Dis 2024; 15:1588-1601. [PMID: 37815906 PMCID: PMC11272210 DOI: 10.14336/ad.2023.0923] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 09/23/2023] [Indexed: 10/12/2023] Open
Abstract
Aging is associated with changes in the immune system and the gut microbiota. Immunosenescence may lead to a low-grade, sterile chronic inflammation in a multifactorial and dynamic way, which plays a critical role in most age-related diseases. Age-related changes in the gut microbiota also shape the immune and inflammatory responses. Nutrition is a determinant of immune function and of the gut microbiota. Immunonutrion has been regarded as a new strategy for disease prevention and management, including many age-related diseases. However, the understanding of the cause-effect relationship is required to be more certain about the role of immunonutrition in supporting the immune homeostasis and its clinical relevance in elderly individuals. Herein, we review the remarkable quantitative and qualitative changes during aging that contribute to immunosenescence, inflammaging and microbial dysbiosis, and the effects on late-life health conditions. Furthermore, we discuss the clinical significance of immunonutrition in the treatment of age-related diseases by systematically reviewing its modulation of the immune system and the gut microbiota to clarify the effect of immunonutrition-based interventions on the healthy aging.
Collapse
Affiliation(s)
- Lei Dou
- Department of Geriatrics, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan 430030, China.
- Department of Surgery, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yang Peng
- Department of Geriatrics, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Bin Zhang
- Department of Surgery, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Huiyuan Yang
- Department of Surgery, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Kai Zheng
- Department of Geriatrics, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
8
|
Cyr B, Curiel Cid R, Loewenstein D, Vontell RT, Dietrich WD, Keane RW, de Rivero Vaccari JP. The Inflammasome Adaptor Protein ASC in Plasma as a Biomarker of Early Cognitive Changes. Int J Mol Sci 2024; 25:7758. [PMID: 39063000 PMCID: PMC11276719 DOI: 10.3390/ijms25147758] [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: 05/31/2024] [Revised: 07/13/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024] Open
Abstract
Dementia is a group of symptoms including memory loss, language difficulties, and other types of cognitive and functional impairments that affects 57 million people worldwide, with the incidence expected to double by 2040. Therefore, there is an unmet need to develop reliable biomarkers to diagnose early brain impairments so that emerging interventions can be applied before brain degeneration. Here, we performed biomarker analyses for apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), and amyloid-β 42/40 (Aβ42/40) ratio in the plasma of older adults. Participants had blood drawn at baseline and underwent two annual clinical and cognitive evaluations. The groups tested either cognitively normal on both evaluations (NN), cognitively normal year 1 but cognitively impaired year 2 (NI), or cognitively impaired on both evaluations (II). ASC was elevated in the plasma of the NI group compared to the NN and II groups. Additionally, Aβ42 was increased in the plasma in the NI and II groups compared to the NN group. Importantly, the area under the curve (AUC) for ASC in participants older than 70 years old in NN vs. NI groups was 0.81, indicating that ASC is a promising plasma biomarker for early detection of cognitive decline.
Collapse
Affiliation(s)
- Brianna Cyr
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA; (B.C.); (W.D.D.); (R.W.K.)
| | - Rosie Curiel Cid
- Center for Cognitive Neuroscience and Aging, University of Miami, Miami, FL 33136, USA; (R.C.C.); (D.L.)
| | - David Loewenstein
- Center for Cognitive Neuroscience and Aging, University of Miami, Miami, FL 33136, USA; (R.C.C.); (D.L.)
| | | | - W. Dalton Dietrich
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA; (B.C.); (W.D.D.); (R.W.K.)
| | - Robert W. Keane
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA; (B.C.); (W.D.D.); (R.W.K.)
- Department of Physiology and Biophysics, University of Miami, Miami, FL 33136, USA
| | - Juan Pablo de Rivero Vaccari
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA; (B.C.); (W.D.D.); (R.W.K.)
- Center for Cognitive Neuroscience and Aging, University of Miami, Miami, FL 33136, USA; (R.C.C.); (D.L.)
- Department of Physiology and Biophysics, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
9
|
Hand LK, Taylor MK, Sullivan DK, Siengsukon CF, Morris JK, Martin LE, Hull HR. Pregnancy as a window of opportunity for dementia prevention: a narrative review. Nutr Neurosci 2024:1-13. [PMID: 38970804 DOI: 10.1080/1028415x.2024.2371727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2024]
Abstract
Dementia is a debilitating condition with a disproportionate impact on women. While sex differences in longevity contribute to the disparity, the role of the female sex as a biological variable in disease progression is not yet fully elucidated. Metabolic dysfunctions are drivers of dementia etiology, and cardiometabolic diseases are among the most influential modifiable risk factors. Pregnancy is a time of enhanced vulnerability for metabolic disorders. Many dementia risk factors, such as hypertension or blood glucose dysregulation, often emerge for the first time in pregnancy. While such cardiometabolic complications in pregnancy pose a risk to the health trajectory of a woman, increasing her odds of developing type 2 diabetes or chronic hypertension, it is not fully understood how this relates to her risk for dementia. Furthermore, structural and functional changes in the maternal brain have been reported during pregnancy suggesting it is a time of neuroplasticity for the mother. Therefore, pregnancy may be a window of opportunity to optimize metabolic health and support the maternal brain. Healthy dietary patterns are known to reduce the risk of cardiometabolic diseases and have been linked to dementia prevention, yet interventions targeting cognitive function in late life have largely been unsuccessful. Earlier interventions are needed to address the underlying metabolic dysfunctions and potentially reduce the risk of dementia, and pregnancy offers an ideal opportunity to intervene. This review discusses current evidence regarding maternal brain health and the potential window of opportunity in pregnancy to use diet to address neurological health disparities for women.
Collapse
Affiliation(s)
- Lauren K Hand
- Department of Dietetics and Nutrition, School of Health Professions, University of Kansas Medical Center, Kansas City, KS, USA
| | - Matthew K Taylor
- Department of Dietetics and Nutrition, School of Health Professions, University of Kansas Medical Center, Kansas City, KS, USA
| | - Debra K Sullivan
- Department of Dietetics and Nutrition, School of Health Professions, University of Kansas Medical Center, Kansas City, KS, USA
| | - Catherine F Siengsukon
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jill K Morris
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Laura E Martin
- Department of Population Health, University of Kansas Medical Center, Kansas City, KS, USA
- Hoglund Biomedical Imaging Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Holly R Hull
- Department of Dietetics and Nutrition, School of Health Professions, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
10
|
Li C, Qian H, Feng L, Li M. Causal Association Between Type 2 Diabetes Mellitus and Alzheimer's Disease: A Two-Sample Mendelian Randomization Study. J Alzheimers Dis Rep 2024; 8:945-957. [PMID: 39114544 PMCID: PMC11305840 DOI: 10.3233/adr-240053] [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: 03/25/2024] [Accepted: 05/17/2024] [Indexed: 08/10/2024] Open
Abstract
Background There is now increasing evidence that type 2 diabetes mellitus (T2DM) is associated with Alzheimer's disease (AD). However, it is unclear whether the two are causally related. Objective To reveal the causal association between T2DM and AD, we performed a bidirectional Mendelian randomization (MR) analysis. Methods Genetic instrumental variables were systematically screened, and inverse-variance weighting, MR-Egger regression, weighted median, simple mode, and weighted mode were applied to assess the pathogenic associations between the two diseases, and sensitivity analyses were used to further validate the robustness of the results. Results The results of forward MR analysis with T2DM as the exposure were [OR = 0.998, 95% CI (0.975∼1.021), p = 0.857], and the results of reverse MR analysis with AD as the exposure were [OR = 0.966, 95% CI (0.934∼0.999), p = 0.043]. The results showed no significant association between T2DM and AD at the gene level (p < 0.025). Sensitivity analyses were consistent with the results of the main analysis, confirming the robustness of the study. Conclusions T2DM and AD may not be genetically causally associated.
Collapse
Affiliation(s)
- Cong Li
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Haifeng Qian
- Department of Image Center, the Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, China
| | - Lina Feng
- Department of Neurology, the Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Mingquan Li
- Neurology Department, the Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
11
|
Lee JY, Wong CY, Koh RY, Lim CL, Kok YY, Chye SM. Natural Bioactive Compounds from Macroalgae and Microalgae for the Treatment of Alzheimer's Disease: A Review. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2024; 97:205-224. [PMID: 38947104 PMCID: PMC11202106 DOI: 10.59249/jnkb9714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Neuroinflammation, toxic protein aggregation, oxidative stress, and mitochondrial dysfunction are key pathways in neurodegenerative diseases like Alzheimer's disease (AD). Targeting these mechanisms with antioxidants, anti-inflammatory compounds, and inhibitors of Aβ formation and aggregation is crucial for treatment. Marine algae are rich sources of bioactive compounds, including carbohydrates, phenolics, fatty acids, phycobiliproteins, carotenoids, fatty acids, and vitamins. In recent years, they have attracted interest from the pharmaceutical and nutraceutical industries due to their exceptional biological activities, which include anti-inflammation, antioxidant, anticancer, and anti-apoptosis properties. Multiple lines of evidence have unveiled the potential neuroprotective effects of these multifunctional algal compounds for application in treating and managing AD. This article will provide insight into the molecular mechanisms underlying the neuroprotective effects of bioactive compounds derived from algae based on in vitro and in vivo models of neuroinflammation and AD. We will also discuss their potential as disease-modifying and symptomatic treatment strategies for AD.
Collapse
Affiliation(s)
- Jia Yee Lee
- School of Health Sciences, International Medical
University, Kuala Lumpur, Malaysia
| | - Chiew Yen Wong
- Department of Applied Biomedical Science and
Biotechnology, School of Health Sciences, International Medical University,
Kuala Lumpur, Malaysia
| | - Rhun Yian Koh
- Department of Applied Biomedical Science and
Biotechnology, School of Health Sciences, International Medical University,
Kuala Lumpur, Malaysia
| | - Chooi Ling Lim
- Department of Applied Biomedical Science and
Biotechnology, School of Health Sciences, International Medical University,
Kuala Lumpur, Malaysia
| | - Yih Yih Kok
- Department of Applied Biomedical Science and
Biotechnology, School of Health Sciences, International Medical University,
Kuala Lumpur, Malaysia
| | - Soi Moi Chye
- Department of Applied Biomedical Science and
Biotechnology, School of Health Sciences, International Medical University,
Kuala Lumpur, Malaysia
| |
Collapse
|
12
|
Wang F, Wang A, Huang Y, Gao W, Xu Y, Zhang W, Guo G, Song W, Kong Y, Wang Q, Wang S, Shi F. Lipoproteins and metabolites in diagnosing and predicting Alzheimer's disease using machine learning. Lipids Health Dis 2024; 23:152. [PMID: 38773573 PMCID: PMC11107010 DOI: 10.1186/s12944-024-02141-w] [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: 02/15/2024] [Accepted: 05/09/2024] [Indexed: 05/24/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a chronic neurodegenerative disorder that poses a substantial economic burden. The Random forest algorithm is effective in predicting AD; however, the key factors influencing AD onset remain unclear. This study aimed to analyze the key lipoprotein and metabolite factors influencing AD onset using machine-learning methods. It provides new insights for researchers and medical personnel to understand AD and provides a reference for the early diagnosis, treatment, and early prevention of AD. METHODS A total of 603 participants, including controls and patients with AD with complete lipoprotein and metabolite data from the Alzheimer's disease Neuroimaging Initiative (ADNI) database between 2005 and 2016, were enrolled. Random forest, Lasso regression, and CatBoost algorithms were employed to rank and filter 213 lipoprotein and metabolite variables. Variables with consistently high importance rankings from any two methods were incorporated into the models. Finally, the variables selected from the three methods, with the participants' age, sex, and marital status, were used to construct a random forest predictive model. RESULTS Fourteen lipoprotein and metabolite variables were screened using the three methods, and 17 variables were included in the AD prediction model based on age, sex, and marital status of the participants. The optimal random forest modeling was constructed with "mtry" set to 3 and "ntree" set to 300. The model exhibited an accuracy of 71.01%, a sensitivity of 79.59%, a specificity of 65.28%, and an AUC (95%CI) of 0.724 (0.645-0.804). When Mean Decrease Accuracy and Gini were used to rank the proteins, age, phospholipids to total lipids ratio in intermediate-density lipoproteins (IDL_PL_PCT), and creatinine were among the top five variables. CONCLUSIONS Age, IDL_PL_PCT, and creatinine levels play crucial roles in AD onset. Regular monitoring of lipoproteins and their metabolites in older individuals is significant for early AD diagnosis and prevention.
Collapse
Affiliation(s)
- Fenglin Wang
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Aimin Wang
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Yiming Huang
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Wenfeng Gao
- Department of Rheumatology and Immunology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261031, China
| | - Yaqi Xu
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Wenjing Zhang
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Guiya Guo
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Wangchen Song
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Yujia Kong
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Qinghua Wang
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Suzhen Wang
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China.
| | - Fuyan Shi
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China.
| |
Collapse
|
13
|
Kullenberg H, Rossen J, Johansson UB, Hagströmer M, Nyström T, Kumlin M, Svedberg MM. Correlations between insulin-degrading enzyme and metabolic markers in patients diagnosed with type 2 diabetes, Alzheimer's disease, and healthy controls: a comparative study. Endocrine 2024; 84:450-458. [PMID: 37980298 PMCID: PMC11076361 DOI: 10.1007/s12020-023-03603-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/07/2023] [Indexed: 11/20/2023]
Abstract
PURPOSE This study aimed to explore correlations between insulin-degrading enzyme (IDE) and markers of metabolic function in a group of patients diagnosed with type 2 diabetes mellitus (T2DM) or Alzheimer's disease (AD) and metabolically healthy volunteers. METHOD We included 120 individuals (47 with T2DM, 9 with AD, and 64 healthy controls). Serum levels of IDE were measured with commercial kits for ELISA. Differences in IDE levels between groups were analyzed with non-parametric ANCOVA, and correlations were analyzed with Spearman's rank correlations. We also investigated the influence of age, sex, and the use of insulin on the correlation using a non-parametric version of partial correlation. RESULTS Patients diagnosed with T2DM had higher IDE levels than patients diagnosed with AD and healthy controls after adjustment for age and sex. IDE was increasingly associated with body mass index (BMI), fasting blood glucose, C-peptide, hemoglobin A1c (HbA1c), insulin resistance, and triglycerides. In stratified analyses, we found a decreasing partial correlation between IDE and HbA1c in patients diagnosed with AD and a decreasing partial correlation between IDE and C-peptide in healthy controls. In patients diagnosed with T2DM, we found no partial correlations. CONCLUSION These results indicate that IDE is essential in metabolic function and might reflect metabolic status, although it is not yet a biomarker that can be utilized in clinical practice. Further research on IDE in human blood may provide crucial insights into the full function of the enzyme.
Collapse
Affiliation(s)
- Helena Kullenberg
- Department of Health Promoting Science, Sophiahemmet University, Stockholm, Sweden.
| | - Jenny Rossen
- Department of Health Promoting Science, Sophiahemmet University, Stockholm, Sweden
| | - Unn-Britt Johansson
- Department of Health Promoting Science, Sophiahemmet University, Stockholm, Sweden
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Stockholm, Sweden
| | - Maria Hagströmer
- Department of Health Promoting Science, Sophiahemmet University, Stockholm, Sweden
- Division of Physiotherapy, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Academic Primary Health Care Centre, Region Stockholm, Stockholm, Sweden
| | - Thomas Nyström
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Stockholm, Sweden
| | - Maria Kumlin
- Department of Health Promoting Science, Sophiahemmet University, Stockholm, Sweden
| | - Marie M Svedberg
- Department of Health Promoting Science, Sophiahemmet University, Stockholm, Sweden
| |
Collapse
|
14
|
Liu ZL, Hua FF, Qu L, Yan N, Zhang HF. Evaluating serum CXCL12, sCD22, Lp-PLA2 levels and ratios as biomarkers for diagnosis of Alzheimer's disease. World J Psychiatry 2024; 14:380-387. [PMID: 38617987 PMCID: PMC11008386 DOI: 10.5498/wjp.v14.i3.380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/15/2024] [Accepted: 02/04/2024] [Indexed: 03/19/2024] Open
Abstract
BACKGROUND Grasping the underlying mechanisms of Alzheimer's disease (AD) is still a work in progress, and existing diagnostic techniques encounter various obstacles. Therefore, the discovery of dependable biomarkers is essential for early detection, tracking the disease's advancement, and steering treatment strategies. AIM To explore the diagnostic potential of serum CXCL12, sCD22, Lp-PLA2, and their ratios in AD, aiming to enhance early detection and inform targeted treatment strategies. METHODS The study was conducted in Dongying people's Hospital from January 2021 to December 2022. Participants included 60 AD patients (AD group) and 60 healthy people (control group). Using a prospective case-control design, the levels of CXCL12, sCD22 and Lp-PLA2 and their ratios were detected by enzyme-linked immunosorbent assay kit in the diagnosis of AD. The differences between the two groups were analyzed by statistical methods, and the corresponding ratio was constructed to improve the specificity and sensitivity of diagnosis. RESULTS Serum CXCL12 levels were higher in the AD group (47.2 ± 8.5 ng/mL) than the control group (32.8 ± 5.7 ng/mL, P < 0.001), while sCD22 levels were lower (14.3 ± 2.1 ng/mL vs 18.9 ± 3.4 ng/mL, P < 0.01). Lp-PLA2 levels were also higher in the AD group (112.5 ± 20.6 ng/mL vs 89.7 ± 15.2 ng/mL, P < 0.05). Significant differences were noted in CXCL12/sCD22 (3.3 vs 1.7, P < 0.001) and Lp-PLA2/sCD22 ratios (8.0 vs 5.2, P < 0.05) between the groups. Receiver operating characteristic analysis confirmed high sensitivity and specificity of these markers and their ratios in distinguishing AD, with area under the curves ranging from 0.568 to 0.787. CONCLUSION Serum CXCL12 and Lp-PLA2 levels were significantly increased, while sCD22 were significantly decreased, as well as increases in the ratios of CXCL12/sCD22 and Lp-PLA2/sCD22, are closely related to the onset of AD. These biomarkers and their ratios can be used as potential diagnostic indicators for AD, providing an important clinical reference for early intervention and treatment.
Collapse
Affiliation(s)
- Zeng-Ling Liu
- Department of Neurology, Dongying People's Hospital, Dongying 257000, Shandong Province, China
| | - Fei-Fei Hua
- Department of Neurology, Dongying People's Hospital, Dongying 257000, Shandong Province, China
| | - Lei Qu
- Department of Neurology, Dongying People's Hospital, Dongying 257000, Shandong Province, China
| | - Na Yan
- Department of Neurology, Dongying People's Hospital, Dongying 257000, Shandong Province, China
| | - Hui-Fang Zhang
- Department of Neurology, Dongying People's Hospital, Dongying 257000, Shandong Province, China
| |
Collapse
|
15
|
Vande Casteele T, Laroy M, Van Cauwenberge M, Koole M, Dupont P, Sunaert S, Van den Stock J, Bouckaert F, Van Laere K, Emsell L, Vandenbulcke M. Preliminary evidence for preserved synaptic density in late-life depression. Transl Psychiatry 2024; 14:145. [PMID: 38485934 PMCID: PMC10940592 DOI: 10.1038/s41398-024-02837-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 02/11/2024] [Accepted: 02/14/2024] [Indexed: 03/18/2024] Open
Abstract
Late-life depression has been consistently associated with lower gray matter volume, the origin of which remains largely unexplained. Recent in-vivo PET findings in early-onset depression and Alzheimer's Disease suggest that synaptic deficits contribute to the pathophysiology of these disorders and may therefore contribute to lower gray matter volume in late-life depression. Here, we investigate synaptic density in vivo for the first time in late-life depression using the synaptic vesicle glycoprotein 2A receptor radioligand 11C-UCB-J. We included 24 currently depressed adults with late-life depression (73.0 ± 6.2 years, 16 female, geriatric depression scale = 19.5 ± 6.8) and 36 age- and gender-matched healthy controls (70.4 ± 6.2 years, 21 female, geriatric depression scale = 2.7 ± 2.9) that underwent simultaneous 11C-UCB-J positron emission tomography (PET) and 3D T1- and T2-FLAIR weighted magnetic resonance (MR) imaging on a 3-tesla PET-MR scanner. We used analyses of variance to test for 11C-UCB-J binding and gray matter volumes differences in regions implicated in depression. The late-life depression group showed a trend in lower gray matter volumes in the hippocampus (p = 0.04), mesial temporal (p = 0.02) and prefrontal cortex (p = 0.02) compared to healthy control group without surviving correction for multiple comparison. However, no group differences in 11C-UCB-J binding were found in these regions nor were any associations between 11C-UCB-J and depressive symptoms. Our data suggests that, in contrast to Alzheimer's Disease, lower gray matter volume in late-life depression is not associated with synaptic density changes. From a therapeutic standpoint, preserved synaptic density in late-life depression may be an encouraging finding.
Collapse
Affiliation(s)
- Thomas Vande Casteele
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, B-3000, Leuven, Belgium.
| | - Maarten Laroy
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, B-3000, Leuven, Belgium
| | - Margot Van Cauwenberge
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, B-3000, Leuven, Belgium
- Neurology, University Hospitals Leuven, B-3000, Leuven, Belgium
| | - Michel Koole
- KU Leuven, Leuven Brain Institute, Department of Imaging and Pathology, Nuclear Medicine, B-3000, Leuven, Belgium
| | - Patrick Dupont
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Laboratory for Cognitive Neurology, B-3000, Leuven, Belgium
| | - Stefan Sunaert
- KU Leuven, Leuven Brain Institute, Department of Imaging and Pathology, Translational MRI, B-3000, Leuven, Belgium
- Radiology, University Hospitals Leuven, B-3000, Leuven, Belgium
| | - Jan Van den Stock
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, B-3000, Leuven, Belgium
- Geriatric Psychiatry, University Psychiatric Center KU Leuven, B-3000, Leuven, Belgium
| | - Filip Bouckaert
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, B-3000, Leuven, Belgium
- Geriatric Psychiatry, University Psychiatric Center KU Leuven, B-3000, Leuven, Belgium
| | - Koen Van Laere
- KU Leuven, Leuven Brain Institute, Department of Imaging and Pathology, Nuclear Medicine, B-3000, Leuven, Belgium
- Nuclear Medicine, University Hospitals Leuven, B-3000, Leuven, Belgium
| | - Louise Emsell
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, B-3000, Leuven, Belgium
- KU Leuven, Leuven Brain Institute, Department of Imaging and Pathology, Translational MRI, B-3000, Leuven, Belgium
- Geriatric Psychiatry, University Psychiatric Center KU Leuven, B-3000, Leuven, Belgium
| | - Mathieu Vandenbulcke
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, B-3000, Leuven, Belgium
- Geriatric Psychiatry, University Psychiatric Center KU Leuven, B-3000, Leuven, Belgium
| |
Collapse
|
16
|
Tang AS, Rankin KP, Cerono G, Miramontes S, Mills H, Roger J, Zeng B, Nelson C, Soman K, Woldemariam S, Li Y, Lee A, Bove R, Glymour M, Aghaeepour N, Oskotsky TT, Miller Z, Allen IE, Sanders SJ, Baranzini S, Sirota M. Leveraging electronic health records and knowledge networks for Alzheimer's disease prediction and sex-specific biological insights. NATURE AGING 2024; 4:379-395. [PMID: 38383858 PMCID: PMC10950787 DOI: 10.1038/s43587-024-00573-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024]
Abstract
Identification of Alzheimer's disease (AD) onset risk can facilitate interventions before irreversible disease progression. We demonstrate that electronic health records from the University of California, San Francisco, followed by knowledge networks (for example, SPOKE) allow for (1) prediction of AD onset and (2) prioritization of biological hypotheses, and (3) contextualization of sex dimorphism. We trained random forest models and predicted AD onset on a cohort of 749 individuals with AD and 250,545 controls with a mean area under the receiver operating characteristic of 0.72 (7 years prior) to 0.81 (1 day prior). We further harnessed matched cohort models to identify conditions with predictive power before AD onset. Knowledge networks highlight shared genes between multiple top predictors and AD (for example, APOE, ACTB, IL6 and INS). Genetic colocalization analysis supports AD association with hyperlipidemia at the APOE locus, as well as a stronger female AD association with osteoporosis at a locus near MS4A6A. We therefore show how clinical data can be utilized for early AD prediction and identification of personalized biological hypotheses.
Collapse
Affiliation(s)
- Alice S Tang
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA.
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, San Francisco and Berkeley, CA, USA.
| | - Katherine P Rankin
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Gabriel Cerono
- Weill Institute for Neuroscience. Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Silvia Miramontes
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Hunter Mills
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Jacquelyn Roger
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Billy Zeng
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Charlotte Nelson
- Weill Institute for Neuroscience. Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Karthik Soman
- Weill Institute for Neuroscience. Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Sarah Woldemariam
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Yaqiao Li
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Albert Lee
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Riley Bove
- Weill Institute for Neuroscience. Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Maria Glymour
- Department of Anesthesiology, Pain, and Perioperative Medicine, Stanford University, Palo Alto, CA, USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Pain, and Perioperative Medicine, Stanford University, Palo Alto, CA, USA
- Department of Pediatrics, Stanford University, Palo Alto, CA, USA
- Department of Biomedical Data Science, Stanford University, Palo Alto, CA, USA
| | - Tomiko T Oskotsky
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Zachary Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Isabel E Allen
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Stephan J Sanders
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Institute of Developmental and Regenerative Medicine, Department of Paediatrics, University of Oxford, Oxford, UK
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Sergio Baranzini
- Weill Institute for Neuroscience. Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA.
- Department of Pediatrics, University of California, San Francisco, CA, USA.
| |
Collapse
|
17
|
Dzhalilova DS, Silina MV, Kosyreva AM, Tsvetkov IS, Makarova OV. Comparative Molecular and Biological Characteristic of the Systemic Inflammatory Response in Adult and Old Male Wistar Rats with Different Resistance to Hypoxia. Bull Exp Biol Med 2024; 176:680-686. [PMID: 38733478 DOI: 10.1007/s10517-024-06090-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Indexed: 05/13/2024]
Abstract
Morphological, molecular, and biological features of the systemic inflammatory response induced by LPS administration were assessed in adult and old male Wistar rats with high and low resistance to hypoxia. In 6 h after LPS administration, mRNA expression levels of Hif1a, Vegf, Nfkb, and level of IL-1β protein in old rats were higher than in adult rats regardless of hypoxia tolerance. The morphometric study showed that the number of neutrophils in the interalveolar septa of the lungs was significantly higher in low-resistant adult and old rats 6 h after LPS administration. Thus, in old male Wistar rats, systemic inflammatory response is more pronounced than in adult rats and depends on the initial tolerance to hypoxia, which should be considered when developing new approaches to the therapy of systemic inflammatory response in individuals of different ages.
Collapse
Affiliation(s)
- D Sh Dzhalilova
- A. P. Avtsyn Research Institute of Human Morphology, A. P. Petrovsky Russian Research Center of Surgery, Moscow, Russia.
| | - M V Silina
- A. P. Avtsyn Research Institute of Human Morphology, A. P. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - A M Kosyreva
- A. P. Avtsyn Research Institute of Human Morphology, A. P. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - I S Tsvetkov
- A. P. Avtsyn Research Institute of Human Morphology, A. P. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - O V Makarova
- A. P. Avtsyn Research Institute of Human Morphology, A. P. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| |
Collapse
|
18
|
Sharma R. Exploring the emerging bidirectional association between inflamm-aging and cellular senescence in organismal aging and disease. Cell Biochem Funct 2024; 42:e3970. [PMID: 38456500 DOI: 10.1002/cbf.3970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/15/2024] [Accepted: 02/27/2024] [Indexed: 03/09/2024]
Abstract
There is strong evidence that most individuals in the elderly population are characterized by inflamm-aging which refers to a subtle increase in the systemic pro-inflammatory environment and impaired innate immune activation. Although a variety of distinct factors are associated with the progression of inflamm-aging, emerging research is demonstrating a dynamic relationship between the processes of cellular senescence and inflamm-aging. Cellular senescence is a recognized factor governing organismal aging, and through a characteristic secretome, accumulating senescent cells can induce and augment a pro-inflammatory tissue environment that provides a rationale for immune system-independent activation of inflamm-aging and associated diseases. There is also accumulating evidence that inflamm-aging or its components can directly accelerate the development of senescent cells and ultimately senescent cell burden in tissues in a likely vicious inflammatory loop. The present review is intended to describe the emerging senescence-based molecular etiology of inflamm-aging as well as the dynamic reciprocal interactions between inflamm-aging and cellular senescence. Therapeutic interventions concurrently targeting cellular senescence and inflamm-aging are discussed and limitations as well as research opportunities have been deliberated. An effort has been made to provide a rationale for integrating inflamm-aging with cellular senescence both as an underlying cause and therapeutic target for further studies.
Collapse
Affiliation(s)
- Rohit Sharma
- Nutrigerontology Laboratory, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, India
| |
Collapse
|
19
|
Karnik SJ, Margetts TJ, Wang HS, Movila A, Oblak AL, Fehrenbacher JC, Kacena MA, Plotkin LI. Mind the Gap: Unraveling the Intricate Dance Between Alzheimer's Disease and Related Dementias and Bone Health. Curr Osteoporos Rep 2024; 22:165-176. [PMID: 38285083 PMCID: PMC10912190 DOI: 10.1007/s11914-023-00847-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 01/30/2024]
Abstract
PURPOSE OF REVIEW This review examines the linked pathophysiology of Alzheimer's disease/related dementia (AD/ADRD) and bone disorders like osteoporosis. The emphasis is on "inflammaging"-a low-level inflammation common to both, and its implications in an aging population. RECENT FINDINGS Aging intensifies both ADRD and bone deterioration. Notably, ADRD patients have a heightened fracture risk, impacting morbidity and mortality, though it is uncertain if fractures worsen ADRD. Therapeutically, agents targeting inflammation pathways, especially Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) and TNF-α, appear beneficial for both conditions. Additionally, treatments like Sirtuin 1 (SIRT-1), known for anti-inflammatory and neuroprotective properties, are gaining attention. The interconnectedness of AD/ADRD and bone health necessitates a unified treatment approach. By addressing shared mechanisms, we can potentially transform therapeutic strategies, enriching our understanding and refining care in our aging society. This review article is part of a series of multiple manuscripts designed to determine the utility of using artificial intelligence for writing scientific reviews.
Collapse
Affiliation(s)
- Sonali J Karnik
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Tyler J Margetts
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Hannah S Wang
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Alexandru Movila
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Adrian L Oblak
- Department of Radiology & Imaging Sciences, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jill C Fehrenbacher
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA.
| | - Lilian I Plotkin
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
20
|
Valentin-Escalera J, Leclerc M, Calon F. High-Fat Diets in Animal Models of Alzheimer's Disease: How Can Eating Too Much Fat Increase Alzheimer's Disease Risk? J Alzheimers Dis 2024; 97:977-1005. [PMID: 38217592 PMCID: PMC10836579 DOI: 10.3233/jad-230118] [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] [Accepted: 11/15/2023] [Indexed: 01/15/2024]
Abstract
High dietary intake of saturated fatty acids is a suspected risk factor for neurodegenerative diseases, including Alzheimer's disease (AD). To decipher the causal link behind these associations, high-fat diets (HFD) have been repeatedly investigated in animal models. Preclinical studies allow full control over dietary composition, avoiding ethical concerns in clinical trials. The goal of the present article is to provide a narrative review of reports on HFD in animal models of AD. Eligibility criteria included mouse models of AD fed a HFD defined as > 35% of fat/weight and western diets containing > 1% cholesterol or > 15% sugar. MEDLINE and Embase databases were searched from 1946 to August 2022, and 32 preclinical studies were included in the review. HFD-induced obesity and metabolic disturbances such as insulin resistance and glucose intolerance have been replicated in most studies, but with methodological variability. Most studies have found an aggravating effect of HFD on brain Aβ pathology, whereas tau pathology has been much less studied, and results are more equivocal. While most reports show HFD-induced impairment on cognitive behavior, confounding factors may blur their interpretation. In summary, despite conflicting results, exposing rodents to diets highly enriched in saturated fat induces not only metabolic defects, but also cognitive impairment often accompanied by aggravated neuropathological markers, most notably Aβ burden. Although there are important variations between methods, particularly the lack of diet characterization, these studies collectively suggest that excessive intake of saturated fat should be avoided in order to lower the incidence of AD.
Collapse
Affiliation(s)
- Josue Valentin-Escalera
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada)
| | - Manon Leclerc
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada)
| | - Frédéric Calon
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada)
| |
Collapse
|
21
|
Gaikwad S, Senapati S, Haque MA, Kayed R. Senescence, brain inflammation, and oligomeric tau drive cognitive decline in Alzheimer's disease: Evidence from clinical and preclinical studies. Alzheimers Dement 2024; 20:709-727. [PMID: 37814508 PMCID: PMC10841264 DOI: 10.1002/alz.13490] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 10/11/2023]
Abstract
Aging, tau pathology, and chronic inflammation in the brain play crucial roles in synaptic loss, neurodegeneration, and cognitive decline in tauopathies, including Alzheimer's disease. Senescent cells accumulate in the aging brain, accelerate the aging process, and promote tauopathy progression through their abnormal inflammatory secretome known as the senescence-associated secretory phenotype (SASP). Tau oligomers (TauO)-the most neurotoxic tau species-are known to induce senescence and the SASP, which subsequently promote neuropathology, inflammation, oxidative stress, synaptic dysfunction, neuronal death, and cognitive dysfunction. TauO, brain inflammation, and senescence are associated with heterogeneity in tauopathy progression and cognitive decline. However, the underlying mechanisms driving the disease heterogeneity remain largely unknown, impeding the development of therapies for tauopathies. Based on clinical and preclinical evidence, this review highlights the critical role of TauO and senescence in neurodegeneration. We discuss key knowledge gaps and potential strategies for targeting senescence and TauO to treat tauopathies. HIGHLIGHTS: Senescence, oligomeric Tau (TauO), and brain inflammation accelerate the aging process and promote the progression of tauopathies, including Alzheimer's disease. We discuss their role in contributing to heterogeneity in tauopathy and cognitive decline. We highlight strategies to target senescence and TauO to treat tauopathies while addressing key knowledge gaps.
Collapse
Affiliation(s)
- Sagar Gaikwad
- The Mitchell Center for Neurodegenerative Diseasesand Department of NeurologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Sudipta Senapati
- The Mitchell Center for Neurodegenerative Diseasesand Department of NeurologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Md. Anzarul Haque
- The Mitchell Center for Neurodegenerative Diseasesand Department of NeurologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Rakez Kayed
- The Mitchell Center for Neurodegenerative Diseasesand Department of NeurologyUniversity of Texas Medical BranchGalvestonTexasUSA
| |
Collapse
|
22
|
Ling M, Gan J, Hu M, Pan F, Liu M. IL1A regulates the inflammation in gout through the Toll-like receptors pathway. Int J Med Sci 2024; 21:188-199. [PMID: 38164346 PMCID: PMC10750337 DOI: 10.7150/ijms.88447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/21/2023] [Indexed: 01/03/2024] Open
Abstract
Objective: Gout is a dangerous metabolic condition related to monosodium urate (MSU). Our aim is to study the molecular mechanisms underlying gout and to identify potential clinical biomarkers by bioinformatics analysis and experimental validation. Methods: In this study, we retrieved the overlapping genes between GSE199950-Differential Expressed Genes (DEGs) dataset and key module in Weighted Gene Co-Expression Network Analysis (WGCNA) on GSE199950. These genes were then analyzed by protein-protein interaction (PPI) network, expression and Gene Set Enrichment Analysis to identify the hub gene related to gout. Then, the gene was investigated by peripheral blood mononuclear cells (PBMCs), immunoassay and cell experiments like western blotting to uncover its underlying mechanism in gout cells. Results: From the turquoise module and 83 DEGs, we identified 62 overlapping genes, only 11 genes had mutual interactions in PPI network and these genes were highly expressed in MSU-treated samples. Then, it was found that the IL1A (interleukin 1 alpha) was the only one gene related to Toll-like receptor signaling pathway that was associated with the occurrence of gout. Thus, IL1A was determined as the hub gene in this study. In immunoassay, IL1A was significantly positively correlated with B cells and negatively correlated with macrophages. Moreover, IL1A is highly expressed in gout patients,it has a good clinical diagnostic value. Finally, the results of in vitro experiments showed that after knocking down IL1A, the expressions of pro-inflammatory cytokines and Toll-like receptor signaling pathway-related proteins (TLR2, TLR4, MyD88) were all reduced. Conclusion: It is confirmed that IL1A is a promoting gene in gout with a good diagnostic value, and specifically it affects the inflammation in gout through Toll-like receptor pathway. Our research offers fresh perspectives on the pathophysiology of gout and valuable directions for future diagnosis and treatment.
Collapse
Affiliation(s)
- Meirong Ling
- Emergency Medical Department, Minhang Hospital, Fudan University, 170 Xinsong Road, 201199, Shanghai, China
| | - Jiaqi Gan
- Department of General Medicine, Minhang Hospital, Fudan University, 170 Xinsong Road, 201199, Shanghai, China
| | - Mengting Hu
- Department of General Medicine, Minhang Hospital, Fudan University, 170 Xinsong Road, 201199, Shanghai, China
| | - Fei Pan
- Department of General Medicine, Minhang Hospital, Fudan University, 170 Xinsong Road, 201199, Shanghai, China
| | - Mei Liu
- Department of General Medicine, Minhang Hospital, Fudan University, 170 Xinsong Road, 201199, Shanghai, China
| |
Collapse
|
23
|
Zhang Y, Tian J, Ni J, Wei M, Li T, Shi J. Peripheral Blood and Cerebrospinal Fluid Levels of YKL-40 in Alzheimer's Disease: A Systematic Review and Meta-Analysis. Brain Sci 2023; 13:1364. [PMID: 37891733 PMCID: PMC10605482 DOI: 10.3390/brainsci13101364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/11/2023] [Accepted: 09/19/2023] [Indexed: 10/29/2023] Open
Abstract
The pathogenesis associated with Alzheimer's disease (AD) is particularly complicated, and early diagnosis and course monitoring of the disease are not ideal based on the available core biomarkers. As a biomarker closely related to neuroinflammation, YKL-40 provides a potential scalable approach in AD, but its association remains controversial and inconclusive with AD. We conducted this study to assess the utility of YKL-40 levels in peripheral blood and cerebrospinal fluid (CSF) of AD patients and healthy controls (HCs) by meta-analysis. We systematically searched and screened relevant trials for comparing YKL-40 levels between AD patients and HCs in PubMed, Embase, Cochrane, and Web of Science, with a search deadline of 14 March 2023 for each database. A total of 17 eligible and relevant studies involving 1811 subjects, including 949 AD patients and 862 HCs, were included. The results showed that YKL-40 levels in the peripheral blood of AD patients and HCs did not possess significant differences. Subgroup analysis showed YKL-40 significantly differed in plasma (SMD = 0.527, 95%CI: [0.302, 0.752]; p = 0.000), but did not in serum. In the case of comparison with HCs, YKL-40 was significantly higher in CSF of AD patients (SMD = 0.893, 95%CI: [0.665, 1.121]; p = 0.000). Besides that, when we performed a combined analysis of total YKL-40 in both peripheral blood and CSF, overall YKL-40 concentrations were also significantly increased among AD patients (SMD = 0.608, 95%CI: [0.272, 0.943]; p = 0.000). YKL-40 provides support and rationale for the neuroinflammatory pathogenesis of AD. The significance of CSF levels of YKL-40 for early screening of AD is definite. Plasma levels of YKL-40 also appear to assist in discriminating AD patients from HCs, which facilitates early screening and monitoring of the natural course of AD.
Collapse
Affiliation(s)
| | | | | | | | | | - Jing Shi
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China; (Y.Z.); (J.T.); (J.N.); (M.W.); (T.L.)
| |
Collapse
|
24
|
Li Y, Wu M, Kong M, Sui S, Wang Q, He Y, Gu J. Impact of Donepezil Supplementation on Alzheimer's Disease-like Pathology and Gut Microbiome in APP/PS1 Mice. Microorganisms 2023; 11:2306. [PMID: 37764150 PMCID: PMC10537997 DOI: 10.3390/microorganisms11092306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/31/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Based on published information, the occurrence and development of Alzheimer's disease (AD) are potentially related to gut microbiota changes. Donepezil hydrochloride (DH), which enhances cholinergic activity by blocking acetylcholinesterase (AChE), is one of the first-line drugs for AD treatment approved by the Food and Drug Administration (FDA) of the USA. However, the potential link between the effects of DH on the pathophysiological processes of AD and the gut microbiota remains unclear. In this study, pathological changes in the brain and colon, the activities of superoxide dismutase (SOD) and AChE, and changes in intestinal flora were observed. The results showed that Aβ deposition in the prefrontal cortex and hippocampus of AD mice was significantly decreased, while colonic inflammation was significantly alleviated by DH treatment. Concomitantly, SOD activity was significantly improved, while AChE was significantly reduced after DH administration. In addition, the gut microbiota community composition of AD mice was significantly altered after DH treatment. The relative abundance of Akkermansia in the AD group was 54.8% higher than that in the N group. The relative abundance of Akkermansia was increased by 18.3% and 53.8% in the AD_G group and the N_G group, respectively. Interestingly, Akkermansia showed a potential predictive value and might be a biomarker for AD. Molecular docking revealed the binding mode and major forces between DH and membrane proteins of Akkermansia. The overall results suggest a novel therapeutic mechanism for treating AD and highlight the critical role of gut microbiota in AD pathology.
Collapse
Affiliation(s)
- Yuan Li
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China; (Y.L.); (M.W.); (M.K.)
| | - Mengyao Wu
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China; (Y.L.); (M.W.); (M.K.)
| | - Mengmeng Kong
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China; (Y.L.); (M.W.); (M.K.)
| | - Shaomei Sui
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China; (S.S.); (Q.W.)
| | - Qi Wang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China; (S.S.); (Q.W.)
| | - Yan He
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China; (S.S.); (Q.W.)
| | - Jinsong Gu
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China; (Y.L.); (M.W.); (M.K.)
| |
Collapse
|
25
|
Dzhalilova DS, Silina MV, Kosyreva AM, Tsvetkov IS, Makarova OV. Morphological and Molecular Biological Features of the Systemic Inflammatory Response in Old Wistar Rats with High and Low Resistance to Hypoxia. Bull Exp Biol Med 2023; 175:704-710. [PMID: 37861901 DOI: 10.1007/s10517-023-05930-y] [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: 02/27/2023] [Indexed: 10/21/2023]
Abstract
Morphological and molecular biological features of LPS-induced systemic inflammatory response were assessed in old male Wistar rats with high (HR) and low (LR) resistance to hypoxia. The systemic inflammatory response was modeled by intraperitoneal injection of E. coli O26:B6 LPS; the animals were sacrificed after 6 h. In histological sections, the number of neutrophils in the interalveolar septa and the area of necrosis in the liver were determined. The expression levels of Hif1a, Hif2a, Nfkb, Vegf, Il1b, Il6, Il10, and Tgfb mRNA in the liver and serum concentrations of HIF-1α and IL-1β were determined. In 4-6 h after LPS injection, 3 (43%) of 7 HR rats died, whereas no deaths were observed among LR rats. At 6 h after LPS injection, the number of neutrophils in the interalveolar septa of the lungs in LR rats was significantly higher than in HR rats, while the area of necrosis in the liver did not differ. At the same time, the mRNA expression levels of the proinflammatory cytokine genes Il1b and Il6 increased in the liver of both HR and LR rats, whereas the expression of Il10 increased only in HR rats. The expression of the Hif1a gene in the liver was higher in LR rats, but the content of HIF-1α protein in blood serum was higher in HR animals. These data should be taken into account when developing new approaches to the therapy of systemic inflammatory response in infectious and inflammatory diseases in the elderly.
Collapse
Affiliation(s)
- D Sh Dzhalilova
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Pet-rovsky National Research Centre of Surgery, Moscow, Russia.
| | - M V Silina
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Pet-rovsky National Research Centre of Surgery, Moscow, Russia
| | - A M Kosyreva
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Pet-rovsky National Research Centre of Surgery, Moscow, Russia
| | - I S Tsvetkov
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Pet-rovsky National Research Centre of Surgery, Moscow, Russia
| | - O V Makarova
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Pet-rovsky National Research Centre of Surgery, Moscow, Russia
| |
Collapse
|
26
|
Sentyabreva AV, Miroshnichenko EA, Melnikova EA, Tsvetkov IS, Kosyreva AM. Morphofunctional Changes in Brain and Peripheral Blood in Adult and Aged Wistar Rats with AlCl 3-Induced Neurodegeneration. Biomedicines 2023; 11:2336. [PMID: 37760778 PMCID: PMC10526012 DOI: 10.3390/biomedicines11092336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/09/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND the general lifespan has been prolonged greatly during the past century, and the incidence of age-associated diseases, including neurodegenerative ones, has increased as well. However, modelling of age-related pathologies is mostly conducted on adult rodents. We studied morphofunctional changes in the brain and peripheral blood of adult Wistar rats in comparison with old Wistar rats to determine age-related physiological changes and differences in adaptive reactions to AlCl3 exposure. METHODS the work was performed on adult and old male Wistar rats. The animals consumed a 100 mg/kg solution of AlCl3 each day for 60 days. Morphological changes of neurons and microglia, mRNA expression levels of pro-inflammatory and anti-inflammatory cytokines, microglia activation markers, amyloid-related proteins, and hallmarks of cellular senescence, monocyte, and lymphocyte subpopulations in the peripheral blood were examined. RESULTS old rats showed increasing hyperchromic neurons in the hippocampus; activation of microglia; upregulation of pro-inflammatory cytokines and cellular senescence markers; downregulation of anti-inflammatory cytokines; and Hif-1a and a decrease in B-cells and monocyte in peripheral blood. CONCLUSION compared to young animals, aged rats respond to aluminum exposure with a severe decline of most cells' function and irreversible neuronal loss. Regarding all reported data, neurodegeneration modelling and investigating of factors capable of accelerating or preventing it should be performed in experimental work on aged animals.
Collapse
Affiliation(s)
- Alexandra Vladislavovna Sentyabreva
- Avtsyn Research Institute of Human Morphology of “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Ekaterina Alexandrovna Miroshnichenko
- Avtsyn Research Institute of Human Morphology of “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Ekaterina Andreevna Melnikova
- Avtsyn Research Institute of Human Morphology of “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
| | - Ivan Sergeevich Tsvetkov
- Avtsyn Research Institute of Human Morphology of “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
| | - Anna Mikhailovna Kosyreva
- Avtsyn Research Institute of Human Morphology of “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| |
Collapse
|
27
|
Kacemi R, Campos MG. Translational Research on Bee Pollen as a Source of Nutrients: A Scoping Review from Bench to Real World. Nutrients 2023; 15:nu15102413. [PMID: 37242296 DOI: 10.3390/nu15102413] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
The emphasis on healthy nutrition is gaining a forefront place in current biomedical sciences. Nutritional deficiencies and imbalances have been widely demonstrated to be involved in the genesis and development of many world-scale public health burdens, such as metabolic and cardiovascular diseases. In recent years, bee pollen is emerging as a scientifically validated candidate, which can help diminish conditions through nutritional interventions. This matrix is being extensively studied, and has proven to be a very rich and well-balanced nutrient pool. In this work, we reviewed the available evidence on the interest in bee pollen as a nutrient source. We mainly focused on bee pollen richness in nutrients and its possible roles in the main pathophysiological processes that are directly linked to nutritional imbalances. This scoping review analyzed scientific works published in the last four years, focusing on the clearest inferences and perspectives to translate cumulated experimental and preclinical evidence into clinically relevant insights. The promising uses of bee pollen for malnutrition, digestive health, metabolic disorders, and other bioactivities which could be helpful to readjust homeostasis (as it is also true in the case of anti-inflammatory or anti-oxidant needs), as well as the benefits on cardiovascular diseases, were identified. The current knowledge gaps were identified, along with the practical challenges that hinder the establishment and fructification of these uses. A complete data collection made with a major range of botanical species allows more robust clinical information.
Collapse
Affiliation(s)
- Rachid Kacemi
- Observatory of Drug-Herb Interactions, Laboratory of Pharmacognosy, Faculty of Pharmacy, University of Coimbra, Heath Sciences Campus, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Maria G Campos
- Observatory of Drug-Herb Interactions, Laboratory of Pharmacognosy, Faculty of Pharmacy, University of Coimbra, Heath Sciences Campus, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra Chemistry Centre (CQC, FCT Unit 313), Faculty of Science and Technology, University of Coimbra, Rua Larga, 3004-516 Coimbra, Portugal
| |
Collapse
|
28
|
Li Y, Chang J, Chen X, Liu J, Zhao L. Advances in the Study of APOE and Innate Immunity in Alzheimer's Disease. J Alzheimers Dis 2023:JAD230179. [PMID: 37182889 DOI: 10.3233/jad-230179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Alzheimer's disease (AD) is a progressive degenerative disease of the nervous system (CNS) with an insidious onset. Clinically, it is characterized by a full range of dementia manifestations including memory impairment, aphasia, loss of speech, loss of use, loss of recognition, impairment of visuospatial skills, and impairment of executive function, as well as changes in personality and behavior. The exact cause of AD has not yet been identified. Nevertheless, modern research indicates that genetic factors contribute to 70% of human's risk of AD. Apolipoprotein (APOE) accounts for up to 90% of the genetic predisposition. APOE is a crucial gene that cannot be overstated. In addition, innate immunity plays a significant role in the etiology and treatment of AD. Understanding the different subtypes of APOE and their interconnections is of paramount importance. APOE and innate immunity, along with their relationship to AD, are primary research motivators for in-depth research and clinical trials. The exploration of novel technologies has led to an increasing trend in the study of AD at the cellular and molecular levels and continues to make more breakthroughs and progress. As of today, there is no effective treatment available for AD around the world. This paper aims to summarize and analyze the role of APOE and innate immunity, as well as development trends in recent years. It is anticipated that APOE and innate immunity will provide a breakthrough for humans to hinder AD progression in the near future.
Collapse
Affiliation(s)
- Yujiao Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jun Chang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xi Chen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jianwei Liu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lan Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
29
|
Chhabra S, Mehan S. Matrine exerts its neuroprotective effects by modulating multiple neuronal pathways. Metab Brain Dis 2023; 38:1471-1499. [PMID: 37103719 DOI: 10.1007/s11011-023-01214-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/10/2023] [Indexed: 04/28/2023]
Abstract
Recent evidence suggests that misfolding, clumping, and accumulation of proteins in the brain may be common causes and pathogenic mechanism for several neurological illnesses. This causes neuronal structural deterioration and disruption of neural circuits. Research from various fields supports this idea, indicating that developing a single treatment for several severe conditions might be possible. Phytochemicals from medicinal plants play an essential part in maintaining the brain's chemical equilibrium by affecting the proximity of neurons. Matrine is a tetracyclo-quinolizidine alkaloid derived from the plant Sophora flavescens Aiton. Matrine has been shown to have a therapeutic effect on Multiple Sclerosis, Alzheimer's disease, and various other neurological disorders. Numerous studies have demonstrated that matrine protects neurons by altering multiple signalling pathways and crossing the blood-brain barrier. As a result, matrine may have therapeutic utility in the treatment of a variety of neurocomplications. This work aims to serve as a foundation for future clinical research by reviewing the current state of matrine as a neuroprotective agent and its potential therapeutic application in treating neurodegenerative and neuropsychiatric illnesses. Future research will answer many concerns and lead to fascinating discoveries that could impact other aspects of matrine.
Collapse
Affiliation(s)
- Swesha Chhabra
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
30
|
Díaz-Camargo E, Hernández-Lalinde J, Sánchez-Rubio M, Chaparro-Suárez Y, Álvarez-Caicedo L, Fierro-Zarate A, Gravini-Donado M, García-Pacheco H, Rojas-Quintero J, Bermúdez V. NHANES 2011-2014 Reveals Decreased Cognitive Performance in U.S. Older Adults with Metabolic Syndrome Combinations. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:5257. [PMID: 37047872 PMCID: PMC10093810 DOI: 10.3390/ijerph20075257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/02/2023] [Accepted: 03/08/2023] [Indexed: 06/19/2023]
Abstract
A relationship between metabolic syndrome and cognitive impairment has been evidenced across research; however, conflicting results have been observed. A cross-sectional study was conducted on 3179 adults older than 60 from the 2011-2014 National Health and Nutrition Examination Survey (NHANES) to analyze the relationship between metabolic syndrome and cognitive impairment. In our results, we found that adults with abdominal obesity, high triglycerides, and low HDL cholesterol had 4.39 fewer points in the CERAD immediate recall test than adults without any metabolic syndrome factors [Beta = -4.39, SE = 1.32, 17.75 (1.36) vs. 22.14 (0.76)]. In addition, people with this metabolic syndrome combination exhibited 2.39 fewer points in the CERAD delayed recall test than those without metabolic syndrome criteria [Beta = -2.39, SE = 0.46, 4.32 (0.49) vs. 6.71 (0.30)]. It was also found that persons with high blood pressure, hyperglycemia, and low HDL-cholesterol levels reached 4.11 points less in the animal fluency test than people with no factors [Beta = -4.11, SE = 1.55, 12.67 (2.12) vs. 16.79 (1.35)]. These findings suggest that specific metabolic syndrome combinations are essential predictors of cognitive impairment. In this study, metabolic syndrome combinations that included obesity, fasting hyperglycemia, high triglycerides, and low HDL-cholesterol were among the most frequent criteria observed.
Collapse
Affiliation(s)
- Edgar Díaz-Camargo
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Cúcuta 540006, Colombia
| | - Juan Hernández-Lalinde
- Facultad de Ciencias Básicas y Biomédicas, Universidad Simón Bolívar, Cúcuta 540006, Colombia
| | - María Sánchez-Rubio
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Cúcuta 540006, Colombia
| | - Yudy Chaparro-Suárez
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Cúcuta 540006, Colombia
| | - Liseth Álvarez-Caicedo
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Cúcuta 540006, Colombia
| | - Alexandra Fierro-Zarate
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Cúcuta 540006, Colombia
| | - Marbel Gravini-Donado
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Barranquilla 080001, Colombia
| | - Henry García-Pacheco
- Facultad de Medicina, Departamento de Cirugía, Universidad del Zulia, Hospital General del Sur, Dr. Pedro Iturbe, Maracaibo 4004, Venezuela
- Unidad de Cirugía para Obesidad y Metabolismo (UCOM), Maracaibo 4004, Venezuela
| | - Joselyn Rojas-Quintero
- Medicine, Pulmonary, Critical Care, and Sleep Medicine Department, Baylor College of Medicine, Houston, TX 77030, USA
| | - Valmore Bermúdez
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla 080001, Colombia
| |
Collapse
|
31
|
Yin T, Liu Y, Ji W, Zhuang J, Chen X, Gong B, Chu J, Liang W, Gao J, Yin Y. Engineered mesenchymal stem cell-derived extracellular vesicles: A state-of-the-art multifunctional weapon against Alzheimer's disease. Theranostics 2023; 13:1264-1285. [PMID: 36923533 PMCID: PMC10008732 DOI: 10.7150/thno.81860] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/21/2023] [Indexed: 02/15/2023] Open
Abstract
With the increase of population aging, the number of Alzheimer's disease (AD) patients is also increasing. According to current estimates, approximately 11% of people over 65 suffer from AD, and that percentage rises to 42% among people over 85. However, no effective treatment capable of decelerating or stopping AD progression is available. Furthermore, AD-targeted drugs composed of synthetic molecules pose concerns regarding biodegradation, clearance, immune response, and neurotoxicity. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) are essential intercellular communication mediators holding great promise as AD therapeutics owing to their biocompatibility, versatility, effortless storage, superior safety, and the ability to transport messenger and noncoding RNAs, proteins, lipids, DNAs, and other bioactive compounds derived from cells. The functionalisation and engineering strategies of MSC-EVs are highlighted (e.g. preconditioning, drug loading, surface modification, and artificial EV fabrication), which could improve AD treatment by multiple therapeutic effects, including clearing abnormal protein accumulation and achieving neuroprotection and immunomodulatory effects. Herein, this review summarises state-of-the-art strategies to engineer MSC-EVs, discusses progress in their use as AD therapeutics, presents the perspectives and challenges associated with the related clinical applications, and concludes that engineered MSC-EVs show immense potential in AD therapy.
Collapse
Affiliation(s)
- Tong Yin
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| | - Yan Liu
- Department of Clinical Pharmacy, Xinhua Hospital; Clinical pharmacy innovation institute, Shanghai Jiao Tong University of Medicine, Shanghai 200000, China
| | - Wenbo Ji
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Jianhua Zhuang
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| | - Xiaohan Chen
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| | - Baofeng Gong
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| | - Jianjian Chu
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| | - Wendanqi Liang
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - You Yin
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| |
Collapse
|
32
|
Grant WB, Blake SM. Diet's Role in Modifying Risk of Alzheimer's Disease: History and Present Understanding. J Alzheimers Dis 2023; 96:1353-1382. [PMID: 37955087 PMCID: PMC10741367 DOI: 10.3233/jad-230418] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2023] [Indexed: 11/14/2023]
Abstract
Diet is an important nonpharmacological risk-modifying factor for Alzheimer's disease (AD). The approaches used here to assess diet's role in the risk of AD include multi-country ecological studies, prospective and cross-sectional observational studies, and laboratory studies. Ecological studies have identified fat, meat, and obesity from high-energy diets as important risk factors for AD and reported that AD rates peak about 15-20 years after national dietary changes. Observational studies have compared the Western dietary pattern with those of the Dietary Approaches to Stop Hypertension (DASH), Mediterranean (MedDi), and Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND) diets. Those studies identified AD risk factors including higher consumption of saturated and total fats, meat, and ultraprocessed foods and a lower risk of AD with higher consumption of fruits, legumes, nuts, omega-3 fatty acids, vegetables, and whole grains. Diet-induced factors associated with a significant risk of AD include inflammation, insulin resistance, oxidative stress, elevated homocysteine, dietary advanced glycation end products, and trimethylamine N-oxide. The molecular mechanisms by which dietary bioactive components and specific foods affect risk of AD are discussed. Given most countries' entrenched food supply systems, the upward trends of AD rates would be hard to reverse. However, for people willing and able, a low-animal product diet with plenty of anti-inflammatory, low-glycemic load foods may be helpful.
Collapse
Affiliation(s)
- William B. Grant
- Sunlight, Nutrition, and Health Research Center, San Francisco, CA, USA
| | - Steven M. Blake
- Nutritional Neuroscience, Maui Memory Clinic, Wailuku, HI, USA
| |
Collapse
|
33
|
Conti E, Grana D, Angiulli F, Karantzoulis A, Villa C, Combi R, Appollonio I, Ferrarese C, Tremolizzo L. TSPO Modulates Oligomeric Amyloid-β-Induced Monocyte Chemotaxis: Relevance for Neuroinflammation in Alzheimer's Disease. J Alzheimers Dis 2023; 95:549-559. [PMID: 37574731 DOI: 10.3233/jad-230239] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
BACKGROUND Neuroinflammation is one of the cardinal mechanisms of Alzheimer's disease (AD). with amyloid-β (Aβ) playing a critical role by activating microglia to produce soluble inflammatory mediators, including several chemokines. Peripheral monocytes are, therefore, attracted into the central nervous system (CNS), where they change into blood-born microglia and participate in the attempt of removing toxic Aβ species. The translocator protein-18 kDa (TSPO) is a transmembrane protein overexpressed in response to neuroinflammation and known to regulate human monocyte chemotaxis. OBJECTIVE We aimed to evaluate the role of the oligomeric Aβ1-42 isoform at inducing peripheral monocyte chemotaxis, and the possible involvement of TSPO in this process. METHODS In vitro cell lines, and ex vivo monocytes from consecutive AD patients (n = 60), and comparable cognitively intact controls (n = 30) were used. Chemotaxis analyses were carried out through both μ-slide chambers and Boyden assays, using 125 pM oligomeric Aβ1-42 as chemoattractant. TSPO agonists and antagonists were tested (Ro5-4864, Emapunil, PK11195). RESULTS Oligomeric Aβ directly promoted chemotaxis in all our models. Interestingly, AD monocytes displayed a stronger response (about twofold) with respect to controls. Aβ-induced chemotaxis was prevented by the TSPO antagonist PK11195; the expression of the TSPO and of the C-C chemokine receptor type 2 (CCR2) was unchanged by drug exposure. CONCLUSION Oligomeric Aβ1-42 is able to recruit peripheral monocytes, and we provide initial evidence sustaining a role for TSPO in modulating this process. This data may be of value for future therapeutic interventions aimed at modulating monocytes motility toward the CNS.
Collapse
Affiliation(s)
- Elisa Conti
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Milan Center for Neuroscience (NeuroMi), Italy
| | - Denise Grana
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Milan Center for Neuroscience (NeuroMi), Italy
| | - Federica Angiulli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Milan Center for Neuroscience (NeuroMi), Italy
| | - Aristotelis Karantzoulis
- Milan Center for Neuroscience (NeuroMi), Italy
- Memory Clinic, Neurology Unit, IRCCS "San Gerardo dei Tintori", Monza, Italy
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Milan Center for Neuroscience (NeuroMi), Italy
| | - Romina Combi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Milan Center for Neuroscience (NeuroMi), Italy
| | - Ildebrando Appollonio
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Milan Center for Neuroscience (NeuroMi), Italy
- Memory Clinic, Neurology Unit, IRCCS "San Gerardo dei Tintori", Monza, Italy
| | - Carlo Ferrarese
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Milan Center for Neuroscience (NeuroMi), Italy
- Memory Clinic, Neurology Unit, IRCCS "San Gerardo dei Tintori", Monza, Italy
| | - Lucio Tremolizzo
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Milan Center for Neuroscience (NeuroMi), Italy
- Memory Clinic, Neurology Unit, IRCCS "San Gerardo dei Tintori", Monza, Italy
| |
Collapse
|