1
|
Qi G, Tang H, Hu J, Kang S, Qin S. Potential role of tanycyte-derived neurogenesis in Alzheimer's disease. Neural Regen Res 2025; 20:1599-1612. [PMID: 38934388 DOI: 10.4103/nrr.nrr-d-23-01865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/17/2024] [Indexed: 06/28/2024] Open
Abstract
Tanycytes, specialized ependymal cells located in the hypothalamus, play a crucial role in the generation of new neurons that contribute to the neural circuits responsible for regulating the systemic energy balance. The precise coordination of the gene networks controlling neurogenesis in naive and mature tanycytes is essential for maintaining homeostasis in adulthood. However, our understanding of the molecular mechanisms and signaling pathways that govern the proliferation and differentiation of tanycytes into neurons remains limited. This article aims to review the recent advancements in research into the mechanisms and functions of tanycyte-derived neurogenesis. Studies employing lineage-tracing techniques have revealed that the neurogenesis specifically originating from tanycytes in the hypothalamus has a compensatory role in neuronal loss and helps maintain energy homeostasis during metabolic diseases. Intriguingly, metabolic disorders are considered early biomarkers of Alzheimer's disease. Furthermore, the neurogenic potential of tanycytes and the state of newborn neurons derived from tanycytes heavily depend on the maintenance of mild microenvironments, which may be disrupted in Alzheimer's disease due to the impaired blood-brain barrier function. However, the specific alterations and regulatory mechanisms governing tanycyte-derived neurogenesis in Alzheimer's disease remain unclear. Accumulating evidence suggests that tanycyte-derived neurogenesis might be impaired in Alzheimer's disease, exacerbating neurodegeneration. Confirming this hypothesis, however, poses a challenge because of the lack of long-term tracing and nucleus-specific analyses of newborn neurons in the hypothalamus of patients with Alzheimer's disease. Further research into the molecular mechanisms underlying tanycyte-derived neurogenesis holds promise for identifying small molecules capable of restoring tanycyte proliferation in neurodegenerative diseases. This line of investigation could provide valuable insights into potential therapeutic strategies for Alzheimer's disease and related conditions.
Collapse
Affiliation(s)
- Guibo Qi
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Han Tang
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jianian Hu
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Siying Kang
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Song Qin
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Siddeeque N, Hussein MH, Abdelmaksoud A, Bishop J, Attia AS, Elshazli RM, Fawzy MS, Toraih EA. Neuroprotective effects of GLP-1 receptor agonists in neurodegenerative Disorders: A Large-Scale Propensity-Matched cohort study. Int Immunopharmacol 2024; 143:113537. [PMID: 39486172 DOI: 10.1016/j.intimp.2024.113537] [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: 09/04/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND GLP-1 receptor agonists, traditionally used for treating type 2 diabetes mellitus and obesity, have demonstrated anti-inflammatory properties. However, their potential neuroprotective effects in neurodegenerative disorders remain unclear. OBJECTIVE To evaluate the impact of GLP-1 receptor agonists on the risk of developing various neurodegenerative conditions in obese patients. METHODS This comprehensive retrospective cohort study analyzed data from 5,307,845 obese adult patients across 73 healthcare organizations in 17 countries. Propensity score matching was performed, resulting in 102,935 patients in each cohort. We compared the risk of developing neurodegenerative disorders between obese patients receiving GLP-1 receptor agonist therapy and those who were not. RESULTS Obese patients treated with GLP-1 receptor agonists showed significantly lower risks of developing Alzheimer's disease (RR = 0.627, 95 %CI = 0.481-0.817), Lewy body dementia (RR = 0.590, 95 %CI = 0.462-0.753), and vascular dementia (RR = 0.438, 95 %CI = 0.327-0.588). The risk reduction for Parkinson's disease was not statistically significant overall (RR = 0.784, 95 %CI = 0.580-1.058) but was significant for semaglutide users (RR = 0.574, 95 %CI = 0.369-0.893). Semaglutide consistently showed the most pronounced protective effects across all disorders. Additionally, a significant reduction in all-cause mortality was observed (HR = 0.525, 95 %CI = 0.493-0.558). CONCLUSION This study provides evidence that the effects of GLP-1 receptor agonists may extend beyond their known metabolic and cardioprotective benefits to include neuroprotection, associated with a decreased risk of developing various neurodegenerative disorders. These findings suggest the potential for expanding the therapeutic applications of GLP-1 receptor agonists to improve neurocognitive outcomes. Further research is warranted to elucidate the mechanisms underlying these neuroprotective effects and to explore their clinical applications in neurodegenerative disease prevention and treatment.
Collapse
Affiliation(s)
| | | | - Ahmed Abdelmaksoud
- Department of Internal Medicine, University of California, Riverside, CA 92521, USA
| | - Julia Bishop
- Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Abdallah S Attia
- Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Rami M Elshazli
- Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA; Biochemistry and Molecular Genetic Unit, Department of Basic Sciences, Faculty of Physical Therapy, Horus University - Egypt, New Damietta 34517, Egypt; Department of Biological Sciences, Faculty of Science, New Mansoura University, New Mansoura City 35742, Egypt
| | - Manal S Fawzy
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar 91431, Saudi Arabia; Center for Health Research, Northern Border University, Arar 1321, Saudi Arabia
| | - Eman A Toraih
- Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA; Medical Genetics Unit, Department of Histology and Cell Biology, Suez Canal University, Ismailia 41522, Egypt.
| |
Collapse
|
3
|
Wong-Guerra M, Montano-Peguero Y, Hernández-Enseñat D, Ramírez-Sánchez J, Mondelo-Rodríguez A, Padrón-Yaquis AS, García-Alfonso E, Fonseca-Fonseca LA, Nuñez-Figueredo Y. Mitochondrial protective properties exerted by JM-20 in a dementia model induced by intracerebroventricular administration of streptozotocin in mice. Behav Brain Res 2024; 480:115385. [PMID: 39667646 DOI: 10.1016/j.bbr.2024.115385] [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: 08/21/2024] [Revised: 11/13/2024] [Accepted: 12/03/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Mitochondrial dysfunction and brain insulin resistance have been related to Alzheimer's disease (AD) development. Streptozotocin (STZ) is commonly employed to disrupt glucose and insulin metabolism, even causing cognitive impairment in animal models. We aimed at studying the protective effect of JM-20 on STZ-induced memory impairment and brain mitochondrial dysfunction. METHODS Male C57Bl6 mice received 3 mg/kg STZ intracerebroventricularly and JM-20 (0.25 mg/kg or 4 mg/kg) was administered daily by gastric gavage. Episodic memory was evaluated through Y-maze, novel object recognition, and Morris water maze. Endogenous antioxidant systems (catalase and superoxide dismutase activities), total sulfhydryl groups, malondialdehyde levels were also studied and acetylcholinesterase (AChE) activity were assessed in the prefrontal cortex (PC) and hippocampus (HO). RESULTS demonstrated that STZ injection impaired recognition and spatial learning and memory and oxygen flow in all mitochondrial respiration states. Additionally, STZ increased AChE, superoxide dismutase, and catalase activity in the PC but not in HO tissue. A neuroprotective effect of JM-20 on STZ-induced memory decline, and mitochondrial dysfunction was observed, suggesting an important causal interaction. In addition, JM-20 was able to decreased AChE enzyme hyperactivity, rescued endogenous antioxidant systems, and prevented histologically observed neuronal damage CONCLUSION: Our results indicate that JM-20 protects against STZ-induced impairment in brain bioenergetic metabolism and memory, confirming its potential as a candidate for treating neurodegenerative disorders associated with mitochondrial dysfunction like AD.
Collapse
Affiliation(s)
- Maylin Wong-Guerra
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos, La Habana 10600, Cuba; Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, USACH, Alameda, Santiago 3363, Chile
| | - Yanay Montano-Peguero
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos, La Habana 10600, Cuba; Facultad de Ciencias Químicas y Farmacéuticas, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santos Dumont 964, Casilla 233, Santiago, Chile
| | - Daniela Hernández-Enseñat
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos, La Habana 10600, Cuba
| | - Jeney Ramírez-Sánchez
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos, La Habana 10600, Cuba
| | - Abel Mondelo-Rodríguez
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos, La Habana 10600, Cuba
| | - Alejandro Saúl Padrón-Yaquis
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos, La Habana 10600, Cuba
| | - Enrique García-Alfonso
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos, La Habana 10600, Cuba
| | - Luis Arturo Fonseca-Fonseca
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos, La Habana 10600, Cuba.
| | - Yanier Nuñez-Figueredo
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos, La Habana 10600, Cuba.
| |
Collapse
|
4
|
Lee WJ, Cho KJ, Kim GW. Mitigation of Atherosclerotic Vascular Damage and Cognitive Improvement Through Mesenchymal Stem Cells in an Alzheimer's Disease Mouse Model. Int J Mol Sci 2024; 25:13210. [PMID: 39684920 DOI: 10.3390/ijms252313210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition characterized by progressive memory loss and other cognitive disturbances. Patients with AD can be vulnerable to vascular damage, and damaged vessels can lead to cognitive impairment. Mesenchymal stem cell (MSC) treatment has shown potential in ameliorating AD pathogenesis, but its effect on vascular function remains unclear. This study aimed to improve cognitive function by alleviating atherosclerosis-induced vessel damage using MSCs in mice with a genetic AD background. In this study, a 5xFAD mouse model of AD was used, and atherosclerotic vessel damage was induced by high-fat diets (HFDs). MSCs were injected into the tail vein along with mannitol in 5xFAD mice on an HFD. MSCs were detected in the brain, and vascular damage was improved following MSC treatment. Behavioral tests showed that MSCs enhanced cognitive function, as measured by the Y-maze and passive avoidance tests. Additionally, muscle strength measured by the rotarod test was also increased by MSCs in AD mice with vessel damage induced by HFDs. Overall, our results suggest that stem cells can alleviate vascular damage caused by metabolic diseases, including HFDs, and vascular disease in individuals carrying the AD gene. Consequently, this alleviates cognitive decline related to vascular dementia symptoms.
Collapse
Affiliation(s)
- Woong Jin Lee
- Department of Neurology, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| | - Kyoung Joo Cho
- Department of Life Science, Kyonggi University, Suwon 16227, Republic of Korea
| | - Gyung Whan Kim
- Department of Neurology, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
5
|
Olvera-Rojas M, Sewell KR, Karikari TK, Huang H, Oberlin LE, Zeng X, Morris JK, Collins AM, Drake JA, Sutton BP, Kramer AF, Hillman CH, Vidoni ED, Burns JM, Kamboh MI, McAuley E, Marsland AL, Chen Y, Lafferty TK, Sehrawat A, Jakicic JM, Wan L, Kang C, Erickson KI. Influence of medical conditions on the diagnostic accuracy of plasma p-tau217 and p-tau217/Aβ42. Alzheimers Dement 2024. [PMID: 39641408 DOI: 10.1002/alz.14430] [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: 09/09/2024] [Revised: 10/26/2024] [Accepted: 11/01/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION Blood-based biomarkers (BBMs) can enable early detection of brain amyloid beta (Aβ) pathology in cognitively unimpaired individuals. However, the extent to which common medical conditions affect biomarker performance remains unclear. METHODS Participants (n = 348) included individuals without cognitive impairment. We studied how brain Aβ associated with BBMs (Aβ42/40, phosphorylated tau [p-tau] 181 and 217, p-tau217/Aβ42, glial fibrillary acidic protein [GFAP], and neurofilament light [NfL]) and optimal BBM thresholds for predicting brain Aβ positivity and whether they are obscured by the presence of common medical conditions. RESULTS Plasma Aβ42/40, p-tau181, p-tau217, and GFAP, but not NfL, were significantly associated with brain Aβ. P-tau217/Aβ42 showed the best discriminative performance (area under the curve: 0.91). The strength of p-tau217-brain Aβ associations were obscured by diabetes and cardiovascular conditions. DISCUSSION These results suggest BBMs may help detect early Aβ pathology but suggest caution in their use due to common medical conditions that could affect accuracy. HIGHLIGHTS Plasma Aβ42/40, p-tau181, p-tau217, and GFAP but not NfL showed significant associations with brain Aβ. BBMs were more strongly associated with the level of brain Aβ in those without diabetes and cardiovascular conditions. P-tau217/Aβ42 showed the best performance (AUC = 0.91) in discriminating Aβ presence with an optimal cut-off of >1.2, followed by p-tau217 at >0.46 pg/mL, with performance slightly improving when excluding participants with cardiovascular conditions.
Collapse
Affiliation(s)
- Marcos Olvera-Rojas
- Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain
| | - Kelsey R Sewell
- AdventHealth Research Institute, Orlando, Florida, USA
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia
| | - Thomas K Karikari
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Haiqing Huang
- AdventHealth Research Institute, Orlando, Florida, USA
| | - Lauren E Oberlin
- AdventHealth Research Institute, Orlando, Florida, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, New York, USA
| | - Xuemei Zeng
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jill K Morris
- University of Kansas Alzheimer's Disease Research Center, Kansas City, Kansas, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | | | - Jermon A Drake
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Bradley P Sutton
- The Grainger College of Engineering, Bioengineering Department, University of Illinois, Champaign, Illinois, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Arthur F Kramer
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Center for Cognitive and Brain Health, Northeastern University, Boston, Massachusetts, USA
- Department of Psychology, Northeastern University, Boston, Massachusetts, USA
| | - Charles H Hillman
- Center for Cognitive and Brain Health, Northeastern University, Boston, Massachusetts, USA
- Department of Psychology, Northeastern University, Boston, Massachusetts, USA
- Department of Physical Therapy, Movement, & Rehabilitation Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Eric D Vidoni
- University of Kansas Alzheimer's Disease Research Center, Kansas City, Kansas, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jeffrey M Burns
- University of Kansas Alzheimer's Disease Research Center, Kansas City, Kansas, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - M Ilyas Kamboh
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- University of Pittsburgh Department of Epidemiology, Pittsburgh, Pennsylvania, USA
- University of Pittsburgh Department of Human Genetics, Pittsburgh, Pennsylvania, USA
| | - Edward McAuley
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Department of Health and Kinesiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Anna L Marsland
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yijun Chen
- Department of Chemistry, Chevron Science Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tara K Lafferty
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anuradha Sehrawat
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John M Jakicic
- Department of Internal Medicine, Division of Physical Activity and Weight Management, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Lu Wan
- AdventHealth Research Institute, Orlando, Florida, USA
| | - Chaeryon Kang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kirk I Erickson
- AdventHealth Research Institute, Orlando, Florida, USA
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
6
|
Jászberényi M, Thurzó B, Jayakumar AR, Schally AV. The Aggravating Role of Failing Neuropeptide Networks in the Development of Sporadic Alzheimer's Disease. Int J Mol Sci 2024; 25:13086. [PMID: 39684795 DOI: 10.3390/ijms252313086] [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: 10/12/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease imposes an increasing burden on aging Western societies. The disorder most frequently appears in its sporadic form, which can be caused by environmental and polygenic factors or monogenic conditions of incomplete penetrance. According to the authors, in the majority of cases, Alzheimer's disease represents an aggravated form of the natural aging of the central nervous system. It can be characterized by the decreased elimination of amyloid β1-42 and the concomitant accumulation of degradation-resistant amyloid plaques. In the present paper, the dysfunction of neuropeptide regulators, which contributes to the pathophysiologic acceleration of senile dementia, is reviewed. However, in the present review, exclusively those neuropeptides or neuropeptide families are scrutinized, and the authors' investigations into their physiologic and pathophysiologic activities have made significant contributions to the literature. Therefore, the pathophysiologic role of orexins, neuromedins, RFamides, corticotrope-releasing hormone family, growth hormone-releasing hormone, gonadotropin-releasing hormone, ghrelin, apelin, and natriuretic peptides are discussed in detail. Finally, the therapeutic potential of neuropeptide antagonists and agonists in the inhibition of disease progression is discussed here.
Collapse
Affiliation(s)
- Miklós Jászberényi
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
| | - Balázs Thurzó
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
- Emergency Patient Care Unit, Albert Szent-Györgyi Health Centre, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Arumugam R Jayakumar
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Andrew V Schally
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
7
|
Zhang W, Young JI, Gomez L, Schmidt MA, Lukacsovich D, Kunkle BW, Chen X, Martin ER, Wang L. Blood DNA Methylation Signature for Incident Dementia: Evidence from Longitudinal Cohorts. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.03.24316667. [PMID: 39649611 PMCID: PMC11623760 DOI: 10.1101/2024.11.03.24316667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
INTRODUCTION Distinguishing between molecular changes that precede dementia onset and those resulting from the disease is challenging with cross-sectional studies. METHODS We studied blood DNA methylation (DNAm) differences and incident dementia in two large longitudinal cohorts: the Offspring cohort of the Framingham Heart Study (FHS) and the Alzheimer's Disease Neuroimaging Initiative (ADNI) study. We analyzed blood DNAm samples from over 1,000 cognitively unimpaired subjects. RESULTS Meta-analysis identified 44 CpGs and 44 differentially methylated regions consistently associated with time to dementia in both cohorts. Our integrative analysis identified early processes in dementia, such as immune responses and metabolic dysfunction. Furthermore, we developed a Methylation-based Risk Score, which successfully predicted future cognitive decline in an independent validation set, even after accounting for age, sex, APOE ε4, years of education, baseline diagnosis, and baseline MMSE score. DISCUSSION DNA methylation offers a promising source of biomarker for early detection of dementia.
Collapse
Affiliation(s)
- Wei Zhang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Juan I. Young
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lissette Gomez
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Michael A. Schmidt
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - David Lukacsovich
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Brian W. Kunkle
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Xi Chen
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Eden R. Martin
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lily Wang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
8
|
Feng Y, Cheng L, Zhou W, Lu J, Huang H. Metabolic Syndrome and the Risk of Alzheimer's Disease: A Meta-Analysis. Metab Syndr Relat Disord 2024. [PMID: 39558765 DOI: 10.1089/met.2024.0155] [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: 11/20/2024] Open
Abstract
Purpose: The relationship between metabolic syndrome (MetS) and the risk of Alzheimer's disease (AD) remains unclear. This meta-analysis aims to clarify the prospective association between MetS and AD risk and to explore how individual MetS components contribute to this relationship. Methods: Comprehensive searches of MEDLINE, Web of Science, and Embase were conducted up to April 12, 2024. Relevant prospective cohort studies were included. Hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated to assess the associations. A random-effects model was used to incorporate the potential impact of heterogeneity. Findings: Six prospective cohort studies with seven datasets, including 484,994 participants and a follow-up of 3.5 to 13.0 years, were included. The pooled analysis showed no significant association between MetS and AD risk (HR: 0.96, 95% CI: 0.89-1.04, P = 0.37; I2 = 0%). Sensitivity and subgroup analyses confirmed these findings. Individual MetS components exhibited varied effects as follows: abdominal obesity was linked to a reduced AD risk (Risk ratio (RR): 0.70, 95% CI: 0.56-0.88, P = 0.002), whereas high blood pressure (BP) (RR: 1.15, 95% CI: 1.04-1.27, P = 0.007) and hyperglycemia (RR: 1.24, 95% CI: 1.08-1.42, P = 0.002) were associated with an increased risk. Low high-density lipoprotein cholesterol and high triglycerides showed no significant associations. Conclusions: This meta-analysis found no significant overall association between MetS and AD risk. However, specific MetS components, such as abdominal obesity, high BP, and hyperglycemia, may influence AD risk differently.
Collapse
Affiliation(s)
- Yanqiong Feng
- Department of Medical Services, Shanghai Civil Affairs Second Mental Health Center, Shanghai, China
| | - Lili Cheng
- Department of General Practice, Community Health Service Center, Shanghai, China
| | - Weiying Zhou
- Department of General Practice, Shanghai Pudong New Area Zhuqiao Community Health Service Center, Shanghai, China
| | - Jiru Lu
- Department of Medical Services, Shanghai Civil Affairs Second Mental Health Center, Shanghai, China
| | - Huiyu Huang
- Department of Psychological Rehabilitation, Shanghai Nanhui Mental Health Center, Shanghai, China
| |
Collapse
|
9
|
Sighencea MG, Popescu RȘ, Trifu SC. From Fundamentals to Innovation in Alzheimer's Disease: Molecular Findings and Revolutionary Therapies. Int J Mol Sci 2024; 25:12311. [PMID: 39596378 PMCID: PMC11594972 DOI: 10.3390/ijms252212311] [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: 10/07/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD) is a global health concern and the leading cause of dementia in the elderly. The prevalence of this neurodegenerative condition is projected to increase concomitantly with increased life expectancy, resulting in a significant economic burden. With very few FDA-approved disease-modifying drugs available for AD, there is an urgent need to develop new compounds capable of impeding the progression of the disease. Given the unclear etiopathogenesis of AD, this review emphasizes the underlying mechanisms of this condition. It explores not only well-studied aspects, such as the accumulation of Aβ plaques and neurofibrillary tangles, but also novel areas, including glymphatic and lymphatic pathways, microbiota and the gut-brain axis, serotoninergic and autophagy alterations, vascular dysfunction, the metal hypothesis, the olfactory pathway, and oral health. Furthermore, the potential molecular targets arising from all these mechanisms have been reviewed, along with novel promising approaches such as nanoparticle-based therapy, neural stem cell transplantation, vaccines, and CRISPR-Cas9-mediated genome editing techniques. Taking into account the overlap of these various mechanisms, individual and combination therapies emerge as the future direction in the AD strategy.
Collapse
Affiliation(s)
| | - Ramona Ștefania Popescu
- Department of Infectious Diseases, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania;
| | - Simona Corina Trifu
- Department of Psychiatry, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania
| |
Collapse
|
10
|
Ganguly U, Carroll T, Nehrke K, Johnson GVW. Mitochondrial Quality Control in Alzheimer's Disease: Insights from Caenorhabditis elegans Models. Antioxidants (Basel) 2024; 13:1343. [PMID: 39594485 PMCID: PMC11590956 DOI: 10.3390/antiox13111343] [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: 09/12/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder that is classically defined by the extracellular deposition of senile plaques rich in amyloid-beta (Aβ) protein and the intracellular accumulation of neurofibrillary tangles (NFTs) that are rich in aberrantly modified tau protein. In addition to aggregative and proteostatic abnormalities, neurons affected by AD also frequently possess dysfunctional mitochondria and disrupted mitochondrial maintenance, such as the inability to eliminate damaged mitochondria via mitophagy. Decades have been spent interrogating the etiopathogenesis of AD, and contributions from model organism research have aided in developing a more fundamental understanding of molecular dysfunction caused by Aβ and toxic tau aggregates. The soil nematode C. elegans is a genetic model organism that has been widely used for interrogating neurodegenerative mechanisms including AD. In this review, we discuss the advantages and limitations of the many C. elegans AD models, with a special focus and discussion on how mitochondrial quality control pathways (namely mitophagy) may contribute to AD development. We also summarize evidence on how targeting mitophagy has been therapeutically beneficial in AD. Lastly, we delineate possible mechanisms that can work alone or in concert to ultimately lead to mitophagy impairment in neurons and may contribute to AD etiopathology.
Collapse
Affiliation(s)
- Upasana Ganguly
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Trae Carroll
- Department of Pathology, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Keith Nehrke
- Department of Medicine, Nephrology Division, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Gail V. W. Johnson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| |
Collapse
|
11
|
Milenkovic D, Nuthikattu S, Norman JE, Villablanca AC. Single Nuclei Transcriptomics Reveals Obesity-Induced Endothelial and Neurovascular Dysfunction: Implications for Cognitive Decline. Int J Mol Sci 2024; 25:11169. [PMID: 39456952 PMCID: PMC11508525 DOI: 10.3390/ijms252011169] [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: 08/03/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Obesity confers risk for cardiovascular disease and vascular dementia. However, genomic alterations modulated by obesity in endothelial cells in the brain and their relationship to other neurovascular unit (NVU) cells are unknown. We performed single nuclei RNA sequencing (snRNAseq) of the NVU (endothelial cells, astrocytes, microglia, and neurons) from the hippocampus of obese (ob/ob) and wild-type (WT) male mice to characterize obesity-induced transcriptomic changes in a key brain memory center and assessed blood-brain barrier permeability (BBB) by gadolinium-enhanced magnetic resonance imaging (MRI). Ob/ob mice displayed obesity, hyperinsulinemia, and impaired glucose tolerance. snRNAseq profiled 14 distinct cell types and 32 clusters within the hippocampus of ob/ob and WT mice and uncovered differentially expressed genes (DEGs) in all NVU cell types, namely, 4462 in neurons, 1386 in astrocytes, 125 in endothelial cells, and 154 in microglia. Gene ontology analysis identified important biological processes such as angiogenesis in endothelial cells and synaptic trafficking in neurons. Cellular pathway analysis included focal adhesion and insulin signaling, which were common to all NVU cell types. Correlation analysis revealed significant positive correlations between endothelial cells and other NVU cell types. Differentially expressed long non-coding RNAs (lncRNAs) were observed in cells of the NVU-affecting pathways such as TNF and mTOR. BBB permeability showed a trend toward increased signal intensity in ob/ob mice. Taken together, our study provides in-depth insight into the molecular mechanisms underlying cognitive dysfunction in obesity and may have implications for therapeutic gene targeting.
Collapse
Affiliation(s)
- Dragan Milenkovic
- Department of Nutrition, University of California, Davis, CA 95616, USA
| | - Saivageethi Nuthikattu
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, CA 95616, USA; (S.N.); (J.E.N.); (A.C.V.)
| | - Jennifer E. Norman
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, CA 95616, USA; (S.N.); (J.E.N.); (A.C.V.)
| | - Amparo C. Villablanca
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, CA 95616, USA; (S.N.); (J.E.N.); (A.C.V.)
| |
Collapse
|
12
|
Siddiqui AJ, Badraoui R, Alshahrani MM, Snoussi M, Jahan S, Siddiqui MA, Khan A, Sulieman AME, Adnan M. A computational and machine learning approach to identify GPR40-targeting agonists for neurodegenerative disease treatment. PLoS One 2024; 19:e0306579. [PMID: 39378198 PMCID: PMC11481007 DOI: 10.1371/journal.pone.0306579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/19/2024] [Indexed: 10/10/2024] Open
Abstract
The G protein-coupled receptor 40 (GPR40) is known to exert a significant influence on neurogenesis and neurodevelopment within the central nervous system of both humans and rodents. Research findings indicate that the activation of GPR40 by an agonist has been observed to promote the proliferation and viability of hypothalamus cells in the human body. The objective of the present study is to discover new agonist compounds for the GPR40 protein through the utilization of machine learning and pharmacophore-based screening techniques, in conjunction with other computational methodologies such as docking, molecular dynamics simulations, free energy calculations, and investigations of the free energy landscape. In the course of our investigation, we successfully identified five unreported agonist compounds that exhibit robust docking score, displayed stability in ligand RMSD and consistent hydrogen bonding with the receptor in the MD trajectories. Free energy calculations were observed to be higher than control molecule. The measured binding affinities of compounds namely 1, 3, 4, 6 and 10 were -13.9, -13.5, -13.4, -12.9, and -12.1 Kcal/mol, respectively. The identified molecular agonist that has been found can be assessed in terms of its therapeutic efficacy in the treatment of neurological diseases.
Collapse
Affiliation(s)
- Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Ha’il, Ha’il, Saudi Arabia
| | - Riadh Badraoui
- Department of Biology, College of Science, University of Ha’il, Ha’il, Saudi Arabia
| | - Mohammed Merae Alshahrani
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Mejdi Snoussi
- Department of Biology, College of Science, University of Ha’il, Ha’il, Saudi Arabia
| | - Sadaf Jahan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, Saudi Arabia
| | | | - Andleeb Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
- Department of Biosciences, Faculty of Science, Integral University, Lucknow, India
| | | | - Mohd Adnan
- Department of Biology, College of Science, University of Ha’il, Ha’il, Saudi Arabia
| |
Collapse
|
13
|
Li Y, Feng Q, Wang S, Li B, Zheng B, Peng N, Li B, Jiang Y, Liu D, Yang Z, Sha F, Tang J. The association between urinary sodium and the risk of dementia: Evidence from a population-based cohort study. J Affect Disord 2024; 362:518-528. [PMID: 39009316 DOI: 10.1016/j.jad.2024.07.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/14/2024] [Accepted: 07/12/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND Sodium intake reduction is crucial for cardiovascular health, however, its lasting impact on dementia remains unclear. METHODS We included 458,577 UK Biobank participants without dementia at baseline. We estimated 24-h urinary sodium (E24hUNa) using spot urinary parameters and obtained the incidence of all-cause dementia, Alzheimer's disease, and vascular dementia from multiple sources. RESULTS The mean E24hUNa was 3.0 g (1st-99th percentile: 1.5 g-5.1 g). Over a mean follow-up of 13.6 years, 7886 (1.7 %) participants developed all-cause dementia, including 3763 (0.8 %) Alzheimer's disease and 1851 (0.4 %) vascular dementia. In the restricted cubic spline model, we identify a potential cutoff of 3.13 g for E24hUNa, below which each 1 g decrease in E24hUNa was associated with 21 % (95 % confidence interval [CI] 1.11-1.34) higher all-cause dementia risk and 35 % (95 % CI 1.11-1.63) higher vascular dementia risk (P-value <0.001 for non-linearity). The hazard ratios were 1.15 (95 % CI, 1.07-1.24) for all-cause dementia and 1.21 (95 % CI 1.04-1.40) for vascular dementia among individuals with E24hUNa below 3.13 g compared to those with E24hUNa higher than 3.13 g. LIMITATIONS One of the major limitations is the estimation of 24-h urinary sodium with spot urine samples. CONCLUSIONS An E24hUNa level below 3.13 g, equivalent to 3.37 g daily sodium intake, is associated with increased risks of all-cause and vascular dementia. This exploratory study suggests a potential lower limit below which the risk of dementia increases with a lower sodium level. Future studies are necessary to validate our findings.
Collapse
Affiliation(s)
- Ying Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, Guangdong Province, China
| | - Qi Feng
- Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, United Kingdom
| | - Shiyu Wang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, Guangdong Province, China
| | - Bingyu Li
- Department of Government, Shenzhen University, 1066 Xueyuan Avenue, Shenzhen 518055, Guangdong Province, China
| | - Bang Zheng
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Nana Peng
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, Guangdong Province, China
| | - Bingli Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, Guangdong Province, China
| | - Yiwen Jiang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, Guangdong Province, China
| | - Di Liu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, Guangdong Province, China
| | - Zhirong Yang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, Guangdong Province, China; Department of Computational Biology and Medical Big Data, Shenzhen University of Advanced Technology, Shenzhen, China; Primary Care Unit, School of Clinical Medicine, University of Cambridge, Box 111 Cambridge Biomedical Campus, Cambridge CB2 0SP, United Kingdom.
| | - Feng Sha
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, Guangdong Province, China.
| | - Jinling Tang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, Guangdong Province, China; Department of Computational Biology and Medical Big Data, Shenzhen University of Advanced Technology, Shenzhen, China; Clinical Data Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou 510623, Guangdong Province, China; Division of Epidemiology, The JC School of Public Health & Primary Care, The Chinese University of Hong Kong, Room 202, School of Public Health Building, Prince of Wales, Hospital, Shatin, New Territories, Hong Kong, China
| |
Collapse
|
14
|
Gómez-Guijarro MD, Cavero-Redondo I, Saz-Lara A, Pascual-Morena C, Álvarez-Bueno C, Martínez-García I. Intranasal insulin effect on cognitive and/or memory impairment: a systematic review and meta-analysis. Cogn Neurodyn 2024; 18:3059-3073. [PMID: 39555259 PMCID: PMC11564437 DOI: 10.1007/s11571-024-10138-5] [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: 08/30/2023] [Revised: 03/21/2024] [Accepted: 06/02/2024] [Indexed: 11/19/2024] Open
Abstract
Background: Cognitive impairment, characterized by deficits in cognitive functions and loss of delayed and immediate recall, disproportionately affects individuals aged 65 years and older, particularly those with comorbid cardiovascular conditions such as hypertension and diabetes mellitus. Objective: This study aimed to investigate the potential association between intranasal insulin and cognitive and/or memory impairment, with a specific focus on delayed and immediate recall, considering the rising prevalence of cognitive disorders in the aging population. Methodology: Employing a rigorous systematic approach, we conducted a thorough search of MEDLINE, Scopus, the Cochrane database, and Web of Science from inception to November 23, 2022, identifying relevant randomized clinical trials. Our analyses encompassed three key aspects: (i) assessing the impact of intranasal insulin on cognitive impairment, (ii) evaluating its effect on delayed recall, and (iii) examining its influence on immediate recall. Results: Five studies meeting the inclusion criteria were included. The results underscored a statistically significant effect of intranasal insulin on delayed memory (effect size: 1.37; 95% CI: 0.65 to 2.09) and overall cognition (effect size: 0.58; 95% CI: 0.08 to 1.08). However, no statistically significant effect was observed for immediate memory (effect size: 0.48; 95% CI: -0.00 to 0.96). Conclusions: This study provides compelling evidence supporting the significance and efficacy of intranasal insulin in enhancing delayed recall and overall cognition. The observed effects hold promise for potential therapeutic interventions in addressing cognitive deficits associated with aging and comorbid conditions. The findings emphasize the need for further research to elucidate the underlying mechanisms and optimize the application of intranasal insulin in cognitive enhancement strategies. Supplementary Information The online version contains supplementary material available at 10.1007/s11571-024-10138-5.
Collapse
Affiliation(s)
| | - Iván Cavero-Redondo
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Alicia Saz-Lara
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
| | | | - Celia Álvarez-Bueno
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
- Universidad Politécnica y Artística del Paraguay, Asunción, Paraguay
| | | |
Collapse
|
15
|
Sewell KR, Collins AM, Mellow ML, Falck RS, Brown BM, Smith AE, Erickson KI. A Compensatory Role of Physical Activity in the Association Between Sleep and Cognition. Exerc Sport Sci Rev 2024; 52:145-151. [PMID: 39190610 DOI: 10.1249/jes.0000000000000345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
We synthesize evidence investigating the hypothesis that greater engagement in physical activity (PA) may compensate for some of the negative cognitive consequences associated with poor sleep in older adults. Potential mechanistic pathways include glymphatic clearance, influences on depression, and other comorbidities. The evidence base is largely cross-sectional and observational, and further experimental studies are required.
Collapse
Affiliation(s)
| | | | - Maddison L Mellow
- Alliance for Research in Exercise, Nutrition and Activity, Allied Health and Human Performance, University of South Australia, Adelaide, Australia
| | - Ryan S Falck
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | | | - Ashleigh E Smith
- Alliance for Research in Exercise, Nutrition and Activity, Allied Health and Human Performance, University of South Australia, Adelaide, Australia
| | | |
Collapse
|
16
|
Дзгоева ФХ, Екушева ЕВ, Демидова ВВ. [Cognitive impairment in patients with obesity and impaired carbohydrate metabolism (dysglycemia)]. PROBLEMY ENDOKRINOLOGII 2024; 70:75-83. [PMID: 39302867 PMCID: PMC11551794 DOI: 10.14341/probl13389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 09/22/2024]
Abstract
Obesity is a chronic disease, heterogeneous in etiology and clinical manifestations, progressing with a natural course, characterized by excessive deposition of fat mass in the body. This pathological condition has taken on the scale of a global epidemic in recent years, which continues to progress steadily, currently affecting more than 2 billion people worldwide. Due to its heterogeneity, obesity has a negative impact on the work of almost all organs and systems of the body, contributing to the emergence of new concomitant diseases and pathological conditions that significantly worsen the quality of life of these patients. Thus, a close relationship between type 2 diabetes mellitus and cognitive impairment has long been known, as well as with a number of other somatic diseases: coronary heart disease, atherosclerosis, non-alcoholic fatty liver disease, dyslipidemia, malignant neoplasms and other associated pathological conditions against the background of overweight and obesity.Currently, the problem of the relationship of cognitive impairment in patients with overweight or changes in the glycemic profile is very relevant, due to the high prevalence and insufficient study of this issue.
Collapse
Affiliation(s)
- Ф. Х. Дзгоева
- Национальный медицинский исследовательский центр эндокринологии
| | - Е. В. Екушева
- Федеральный научно-клинический центр специализированных видов медицинской помощи и медицинских технологий Федерального медико-биологического агентства; Белгородский государственный национальный исследовательский университет
| | - В. В. Демидова
- Национальный медицинский исследовательский центр эндокринологии
| |
Collapse
|
17
|
Duranti E, Villa C. From Brain to Muscle: The Role of Muscle Tissue in Neurodegenerative Disorders. BIOLOGY 2024; 13:719. [PMID: 39336146 PMCID: PMC11428675 DOI: 10.3390/biology13090719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/02/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024]
Abstract
Neurodegenerative diseases (NDs), like amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), and Parkinson's disease (PD), primarily affect the central nervous system, leading to progressive neuronal loss and motor and cognitive dysfunction. However, recent studies have revealed that muscle tissue also plays a significant role in these diseases. ALS is characterized by severe muscle wasting as a result of motor neuron degeneration, as well as alterations in gene expression, protein aggregation, and oxidative stress. Muscle atrophy and mitochondrial dysfunction are also observed in AD, which may exacerbate cognitive decline due to systemic metabolic dysregulation. PD patients exhibit muscle fiber atrophy, altered muscle composition, and α-synuclein aggregation within muscle cells, contributing to motor symptoms and disease progression. Systemic inflammation and impaired protein degradation pathways are common among these disorders, highlighting muscle tissue as a key player in disease progression. Understanding these muscle-related changes offers potential therapeutic avenues, such as targeting mitochondrial function, reducing inflammation, and promoting muscle regeneration with exercise and pharmacological interventions. This review emphasizes the importance of considering an integrative approach to neurodegenerative disease research, considering both central and peripheral pathological mechanisms, in order to develop more effective treatments and improve patient outcomes.
Collapse
Affiliation(s)
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| |
Collapse
|
18
|
Lim L. Modifying Alzheimer's disease pathophysiology with photobiomodulation: model, evidence, and future with EEG-guided intervention. Front Neurol 2024; 15:1407785. [PMID: 39246604 PMCID: PMC11377238 DOI: 10.3389/fneur.2024.1407785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 08/01/2024] [Indexed: 09/10/2024] Open
Abstract
This manuscript outlines a model of Alzheimer's Disease (AD) pathophysiology in progressive layers, from its genesis to the development of biomarkers and then to symptom expression. Genetic predispositions are the major factor that leads to mitochondrial dysfunction and subsequent amyloid and tau protein accumulation, which have been identified as hallmarks of AD. Extending beyond these accumulations, we explore a broader spectrum of pathophysiological aspects, including the blood-brain barrier, blood flow, vascular health, gut-brain microbiodata, glymphatic flow, metabolic syndrome, energy deficit, oxidative stress, calcium overload, inflammation, neuronal and synaptic loss, brain matter atrophy, and reduced growth factors. Photobiomodulation (PBM), which delivers near-infrared light to selected brain regions using portable devices, is introduced as a therapeutic approach. PBM has the potential to address each of these pathophysiological aspects, with data provided by various studies. They provide mechanistic support for largely small published clinical studies that demonstrate improvements in memory and cognition. They inform of PBM's potential to treat AD pending validation by large randomized controlled studies. The presentation of brain network and waveform changes on electroencephalography (EEG) provide the opportunity to use these data as a guide for the application of various PBM parameters to improve outcomes. These parameters include wavelength, power density, treatment duration, LED positioning, and pulse frequency. Pulsing at specific frequencies has been found to influence the expression of waveforms and modifications of brain networks. The expression stems from the modulation of cellular and protein structures as revealed in recent studies. These findings provide an EEG-based guide for the use of artificial intelligence to personalize AD treatment through EEG data feedback.
Collapse
Affiliation(s)
- Lew Lim
- Vielight Inc., Toronto, ON, Canada
| |
Collapse
|
19
|
Bai M, Huang Z, Zheng X, Hou M, Zhang S. Polysaccharides from Trametes versicolor as a Potential Prebiotic to Improve the Gut Microbiota in High-Fat Diet Mice. Microorganisms 2024; 12:1654. [PMID: 39203496 PMCID: PMC11356736 DOI: 10.3390/microorganisms12081654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 09/03/2024] Open
Abstract
Polysaccharides derived from Trametes versicolor have been found to exhibit hypolipidemic activity in hyperlipidemic mice, but the mechanism by which they modulate intestinal flora is still unclear. Currently, this study aimed to investigate the regulatory effects of extracellular (EPTV) and intracellular polysaccharides from T. versicolor (IPTV) on the dysbiosis of intestinal flora in mice fed a high-fat diet (HFD). The results showed that the oral administration of T. versicolor polysaccharides significantly ameliorated lipid accumulation and steatosis in hepatocytes. The gut dysbiosis in the HFD mice was characterized by a decrease in abundance and diversity of bacteria and an increase in the Firmicutes/Bacteroidetes ratio. However, T. versicolor polysaccharides attenuated these changes and reduced the relative abundance of bile-salt-hydrolase (BSH)-producing bacteria, such as Bacillus, Enterococcus, Bifidobacterium, and Lactococcus. It is noteworthy that T. versicolor polysaccharides also restored the disorganization of intestinal fungi in HFD mice, with EPTV treatment leading to a higher relative abundance of Basidiomycota and Ascomycota compared to IPTV. Additionally, T. versicolor polysaccharides enhanced the growth of butyrate-producing bacteria via the buk and but pathways, accompanied by an increase in short-chain fatty acids (SCFAs), especially butyrate. IPTV also increased the expression of G-protein-coupled receptors 41 (GPR41) and 43 (GPR43) by 40.52% and 113.24% each, as compared to 62.42% and 110.28%, respectively, for EPTV. It is suggested that IPTV and EPTV have the potential to counteract hyperlipidemia-associated intestinal flora disorders and improve lipid metabolism.
Collapse
Affiliation(s)
| | | | | | | | - Song Zhang
- School of Life Science, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
20
|
Wanionok NE, Morel GR, Fernández JM. Osteoporosis and Alzheimer´s disease (or Alzheimer´s disease and Osteoporosis). Ageing Res Rev 2024; 99:102408. [PMID: 38969142 DOI: 10.1016/j.arr.2024.102408] [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: 02/28/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Alzheimer's disease (AD) and osteoporosis are two diseases that mainly affect elderly people, with increases in the occurrence of cases due to a longer life expectancy. Several epidemiological studies have shown a reciprocal association between both diseases, finding an increase in incidence of osteoporosis in patients with AD, and a higher burden of AD in osteoporotic patients. This epidemiological relationship has motivated the search for molecules, genes, signaling pathways and mechanisms that are related to both pathologies. The mechanisms found in these studies can serve to improve treatments and establish better patient care protocols.
Collapse
Affiliation(s)
- Nahuel E Wanionok
- Laboratorio de Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Cs. Exactas. Universidad Nacional de La Plata UNLP-CIC, Argentina
| | - Gustavo R Morel
- Biochemistry Research Institute of La Plata "Professor Doctor Rodolfo R. Brenner" (INIBIOLP), Argentina
| | - Juan M Fernández
- Laboratorio de Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Cs. Exactas. Universidad Nacional de La Plata UNLP-CIC, Argentina.
| |
Collapse
|
21
|
Jung YH, Chae CW, Han HJ. The potential role of gut microbiota-derived metabolites as regulators of metabolic syndrome-associated mitochondrial and endolysosomal dysfunction in Alzheimer's disease. Exp Mol Med 2024; 56:1691-1702. [PMID: 39085351 PMCID: PMC11372123 DOI: 10.1038/s12276-024-01282-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/20/2024] [Accepted: 05/10/2024] [Indexed: 08/02/2024] Open
Abstract
Although the role of gut microbiota (GMB)-derived metabolites in mitochondrial and endolysosomal dysfunction in Alzheimer's disease (AD) under metabolic syndrome remains unclear, deciphering these host-metabolite interactions represents a major public health challenge. Dysfunction of mitochondria and endolysosomal networks (ELNs) plays a crucial role in metabolic syndrome and can exacerbate AD progression, highlighting the need to study their reciprocal regulation for a better understanding of how AD is linked to metabolic syndrome. Concurrently, metabolic disorders are associated with alterations in the composition of the GMB. Recent evidence suggests that changes in the composition of the GMB and its metabolites may be involved in AD pathology. This review highlights the mechanisms of metabolic syndrome-mediated AD development, focusing on the interconnected roles of mitochondrial dysfunction, ELN abnormalities, and changes in the GMB and its metabolites. We also discuss the pathophysiological role of GMB-derived metabolites, including amino acids, fatty acids, other metabolites, and extracellular vesicles, in mediating their effects on mitochondrial and ELN dysfunction. Finally, this review proposes therapeutic strategies for AD by directly modulating mitochondrial and ELN functions through targeting GMB metabolites under metabolic syndrome.
Collapse
Affiliation(s)
- Young Hyun Jung
- Department of Physiology, College of Medicine, Soonchunhyang University, Cheonan, 31151, Korea
| | - Chang Woo Chae
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea.
| |
Collapse
|
22
|
Shippy DC, Evered AH, Ulland TK. Ketone body metabolism and the NLRP3 inflammasome in Alzheimer's disease. Immunol Rev 2024. [PMID: 38989642 DOI: 10.1111/imr.13365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Alzheimer's disease (AD) is a degenerative brain disorder and the most common form of dementia. AD pathology is characterized by senile plaques and neurofibrillary tangles (NFTs) composed of amyloid-β (Aβ) and hyperphosphorylated tau, respectively. Neuroinflammation has been shown to drive Aβ and tau pathology, with evidence suggesting the nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome as a key pathway in AD pathogenesis. NLRP3 inflammasome activation in microglia, the primary immune effector cells of the brain, results in caspase-1 activation and secretion of IL-1β and IL-18. Recent studies have demonstrated a dramatic interplay between the metabolic state and effector functions of immune cells. Microglial metabolism in AD is of particular interest, as ketone bodies (acetone, acetoacetate (AcAc), and β-hydroxybutyrate (BHB)) serve as an alternative energy source when glucose utilization is compromised in the brain of patients with AD. Furthermore, reduced cerebral glucose metabolism concomitant with increased BHB levels has been demonstrated to inhibit NLRP3 inflammasome activation. Here, we review the role of the NLRP3 inflammasome and microglial ketone body metabolism in AD pathogenesis. We also highlight NLRP3 inflammasome inhibition by several ketone body therapies as a promising new treatment strategy for AD.
Collapse
Affiliation(s)
- Daniel C Shippy
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Abigail H Evered
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
- Cellular and Molecular Pathology Graduate Program, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Tyler K Ulland
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
23
|
Yu MH, Lim JS, Yi HA, Won KS, Kim HW. Association between Visceral Adipose Tissue Metabolism and Cerebral Glucose Metabolism in Patients with Cognitive Impairment. Int J Mol Sci 2024; 25:7479. [PMID: 39000586 PMCID: PMC11242271 DOI: 10.3390/ijms25137479] [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: 06/05/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Visceral adipose tissue (VAT) dysfunction has been recently recognized as a potential contributor to the development of Alzheimer's disease (AD). This study aimed to explore the relationship between VAT metabolism and cerebral glucose metabolism in patients with cognitive impairment. This cross-sectional prospective study included 54 patients who underwent 18F-fluorodeoxyglucose (18F-FDG) brain and torso positron emission tomography/computed tomography (PET/CT), and neuropsychological evaluations. VAT metabolism was measured by 18F-FDG torso PET/CT, and cerebral glucose metabolism was measured using 18F-FDG brain PET/CT. A voxel-based analysis revealed that the high-VAT-metabolism group exhibited a significantly lower cerebral glucose metabolism in AD-signature regions such as the parietal and temporal cortices. In the volume-of-interest analysis, multiple linear regression analyses with adjustment for age, sex, and white matter hyperintensity volume revealed that VAT metabolism was negatively associated with cerebral glucose metabolism in AD-signature regions. In addition, higher VAT metabolism was correlated with poorer outcomes on cognitive assessments, including the Korean Boston Naming Test, Rey Complex Figure Test immediate recall, and the Controlled Oral Word Association Test. In conclusion, our study revealed significant relationships among VAT metabolism, cerebral glucose metabolism, and cognitive function. This suggests that VAT dysfunction actively contributes to the neurodegenerative processes characteristic of AD, making VAT dysfunction targeting a novel AD therapy approach.
Collapse
Affiliation(s)
- Mi-Hee Yu
- Department of Nuclear Medicine, Keimyung University Dongsan Hospital, Daegu 42601, Republic of Korea
| | - Ji Sun Lim
- Department of Nuclear Medicine, Keimyung University Dongsan Hospital, Daegu 42601, Republic of Korea
| | - Hyon-Ah Yi
- Department of Neurology, Keimyung University Dongsan Hospital, Daegu 42601, Republic of Korea
| | - Kyoung Sook Won
- Department of Nuclear Medicine, Keimyung University Dongsan Hospital, Daegu 42601, Republic of Korea
| | - Hae Won Kim
- Department of Nuclear Medicine, Keimyung University Dongsan Hospital, Daegu 42601, Republic of Korea
| |
Collapse
|
24
|
Gendron WH, Fertan E, Roddick KM, Wong AA, Maliougina M, Hiani YE, Anini Y, Brown RE. Intranasal insulin treatment ameliorates spatial memory, muscular strength, and frailty deficits in 5xFAD mice. Physiol Behav 2024; 281:114583. [PMID: 38750806 DOI: 10.1016/j.physbeh.2024.114583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
The 5xFAD mouse model shows age-related weight loss as well as cognitive and motor deficits. Metabolic dysregulation, especially impaired insulin signaling, is also present in AD. This study examined whether intranasal delivery of insulin (INI) at low (0.875 U) or high (1.750 U) doses would ameliorate these deficits compared to saline in 10-month-old female 5xFAD and B6SJL wildtype (WT) mice. INI increased forelimb grip strength in the wire hang test in 5xFAD mice in a dose-dependent manner but did not improve the performance of 5xFAD mice on the balance beam. High INI doses reduced frailty scores in 5xFAD mice and improved spatial memory in both acquisition and reversal probe trials in the Morris water maze. INI increased swim speed in 5xFAD mice but had no effect on object recognition memory or working memory in the spontaneous alternation task, nor did it improve memory in the contextual or cued fear memory tasks. High doses of insulin increased the liver, spleen, and kidney weights and reduced brown adipose tissue weights. P-Akt signaling in the hippocampus was increased by insulin in a dose-dependent manner. Altogether, INI increased strength, reduced frailty scores, and improved visual spatial memory. Hypoglycemia was not present after INI, however alterations in tissue and organ weights were present. These results are novel and important as they indicate that intra-nasal insulin can reverse cognitive, motor and frailty deficits found in this mouse model of AD.
Collapse
Affiliation(s)
- William H Gendron
- Departments of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Emre Fertan
- Departments of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Kyle M Roddick
- Departments of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Aimée A Wong
- Departments of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Maria Maliougina
- Departments of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Yassine El Hiani
- Departments of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Younes Anini
- Departments of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada; Departments of Obstetrics and Gynecology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Richard E Brown
- Departments of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada; Departments of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada.
| |
Collapse
|
25
|
Weber CM, Moiz B, Clyne AM. Brain microvascular endothelial cell metabolism and its ties to barrier function. VITAMINS AND HORMONES 2024; 126:25-75. [PMID: 39029976 DOI: 10.1016/bs.vh.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
Brain microvascular endothelial cells, which lie at the interface between blood and brain, are critical to brain energetics. These cells must precisely balance metabolizing nutrients for their own demands with transporting nutrients into the brain to sustain parenchymal cells. It is essential to understand this integrated metabolism and transport so that we can develop better diagnostics and therapeutics for neurodegenerative diseases such as Alzheimer's disease, multiple sclerosis, and traumatic brain injury. In this chapter, we first describe brain microvascular endothelial cell metabolism and how these cells regulate both blood flow and nutrient transport. We then explain the impact of brain microvascular endothelial cell metabolism on the integrity of the blood-brain barrier, as well as how metabolites produced by the endothelial cells impact other brain cells. We detail some ways that cell metabolism is typically measured experimentally and modeled computationally. Finally, we describe changes in brain microvascular endothelial cell metabolism in aging and neurodegenerative diseases. At the end of the chapter, we highlight areas for future research in brain microvascular endothelial cell metabolism. The goal of this chapter is to underscore the importance of nutrient metabolism and transport at the brain endothelium for cerebral health and neurovascular disease treatment.
Collapse
Affiliation(s)
- Callie M Weber
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Bilal Moiz
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Alisa Morss Clyne
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States.
| |
Collapse
|
26
|
Lee S, Byun MS, Yi D, Ahn H, Jung G, Jung JH, Chang YY, Kim K, Choi H, Choi J, Lee JY, Kang KM, Sohn CH, Lee YS, Kim YK, Lee DY. Plasma Leptin and Alzheimer Protein Pathologies Among Older Adults. JAMA Netw Open 2024; 7:e249539. [PMID: 38700863 PMCID: PMC11069086 DOI: 10.1001/jamanetworkopen.2024.9539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 03/01/2024] [Indexed: 05/06/2024] Open
Abstract
Importance Many epidemiologic studies have suggested that low levels of plasma leptin, a major adipokine, are associated with increased risk of Alzheimer disease (AD) dementia and cognitive decline. Nevertheless, the mechanistic pathway linking plasma leptin and AD-related cognitive decline is not yet fully understood. Objective To examine the association of plasma leptin levels with in vivo AD pathologies, including amyloid-beta (Aβ) and tau deposition, through both cross-sectional and longitudinal approaches among cognitively unimpaired older adults. Design, Setting, and Participants This was a longitudinal cohort study from the Korean Brain Aging Study for Early Diagnosis and Prediction of Alzheimer Disease. Data were collected from January 1, 2014, to December 31, 2020, and data were analyzed from July 11 to September 6, 2022. The study included a total of 208 cognitively unimpaired participants who underwent baseline positron emission tomography (PET) scans for brain Aβ deposition. For longitudinal analyses, 192 participants who completed both baseline and 2-year follow-up PET scans for brain Aβ deposition were included. Exposure Plasma leptin levels as assessed by enzyme-linked immunosorbent assay. Main Outcomes and Measures Baseline levels and longitudinal changes of global Aβ and AD-signature region tau deposition measured by PET scans. Results Among the 208 participants, the mean (SD) age was 66.0 (11.3) years, 114 were women (54.8%), and 37 were apolipoprotein E ε4 carriers (17.8%). Lower plasma leptin levels had a significant cross-sectional association with greater brain Aβ deposition (β = -0.04; 95% CI, -0.09 to 0.00; P = .046), while there was no significant association between plasma leptin levels and tau deposition (β = -0.02; 95% CI, -0.05 to 0.02; P = .41). In contrast, longitudinal analyses revealed that there was a significant association between lower baseline leptin levels and greater increase of tau deposition over 2 years (β = -0.06; 95% CI, -0.11 to -0.01; P = .03), whereas plasma leptin levels did not have a significant association with longitudinal change of Aβ deposition (β = 0.006; 95% CI, 0.00-0.02; P = .27). Conclusions and Relevance The present findings suggest that plasma leptin may be protective for the development or progression of AD pathology, including both Aβ and tau deposition.
Collapse
Affiliation(s)
- Seunghoon Lee
- Department of Psychiatry, Myongji Hospital, Hanyang University College of Medicine, Goyang, Republic of Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Hyejin Ahn
- Interdisciplinary Program of Cognitive Science, Seoul National University College of Humanities, Seoul, Republic of Korea
| | - Gijung Jung
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Joon Hyung Jung
- Department of Psychiatry, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Yoon Young Chang
- Department of Psychiatry, Inje University, Sanggye Paik Hospital, Seoul, Republic of Korea
| | - Kyungtae Kim
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyeji Choi
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jeongmin Choi
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jun-Young Lee
- Department of Neuropsychiatry, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
- Interdisciplinary Program of Cognitive Science, Seoul National University College of Humanities, Seoul, Republic of Korea
| |
Collapse
|
27
|
Xu Y, Zhang J, Li X. Erjingwan and Alzheimer's disease: research based on network pharmacology and experimental confirmation. Front Pharmacol 2024; 15:1328334. [PMID: 38741585 PMCID: PMC11089143 DOI: 10.3389/fphar.2024.1328334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Abstract
Background Alzheimer's disease (AD), a challenging neurodegenerative condition, has emerged as a significant global public health concern. The Chinese medicine decoction Erjingwan (EJW) has shown promising efficacy in AD treatment, though its mechanism remains unclear. Objective This study aims to elucidate the mechanism by which EJW treats AD through network pharmacology analysis and in vivo experiments. Methods We identified EJW's components using the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database and determined AD-related targets from various databases. A network comprising herbs-compounds-targets was established, and EJW's core targets were ascertained through protein-protein interaction (PPI) analysis. This study assessed the cognitive abilities of APP/PS1 mice using Morris water mazes and Y mazes, in addition to analyzing blood samples for triglyceride (TG), total cholesterol (TC), low-density lipoprotein (LDL), and high-density lipoprotein (HDL) levels. Brain tissues were examined histologically with HE staining, Nissl staining, and immunohistochemistry (IHC) for amyloid β-protein (Aβ) detection. Superoxide dismutase (SOD), reactive oxygen species (ROS), Interleukin-1β (IL-1β), and Interleukin-6 (IL-6) levels in the hippocampal region were measured by ELISA. mRNA expression of apolipoprotein A-I (APOA-I), apolipoprotein B (APOB), apolipoprotein E4 (APOE4), advanced glycation end products (AGE), the receptor for AGE (RAGE), and nuclear factor kappa-B (NF-κB) was evaluated by quantitative PCR (q-PCR). Western blotting was used to detect the expression of AGE, RAGE, NF-κB, and Tau protein. Results Screening identified 57 chemical components and 222 potential targets of EJW. Ten core targets for AD treatment were identified, with enrichment analysis suggesting EJW's effects are related to lipid metabolism and AGEs/RAGE pathways. EJW enhanced learning and memory in APP/PS1 mice, protected neuronal structure in the hippocampal region, reduced Aβ deposition, and altered levels of TG, TC, LDL, IL-1β, and IL-6, and the expression of APOE4, AGEs, RAGE, NF-κB, and Tau protein, while increasing SOD, APOA-I, and APOB mRNA expression. Conclusion The study identified four core components of EJW-iosgenin, baicalein, beta-sitosterol, quercetin-and ten core targets including AKT1, IL6, VEGFA, TP53, CASP3, for treating AD. Experimental results demonstrate EJW's capacity to modulate lipid profiles, reduce pathological markers such as Aβ1-42, Tau, IL-6, IL-1β, reactive oxygen species, SOD, and enhance cognitive functions in APP/PS1 mice, potentially through inhibiting the AGEs/RAGE/NF-κB pathway.
Collapse
Affiliation(s)
- Yuya Xu
- Department of Neurology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jian Zhang
- School of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Xuling Li
- Department of Neurology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
28
|
Veneziani I, Grimaldi A, Marra A, Morini E, Culicetto L, Marino S, Quartarone A, Maresca G. Towards a Deeper Understanding: Utilizing Machine Learning to Investigate the Association between Obesity and Cognitive Decline-A Systematic Review. J Clin Med 2024; 13:2307. [PMID: 38673581 PMCID: PMC11051247 DOI: 10.3390/jcm13082307] [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: 03/18/2024] [Revised: 04/09/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Background/Objectives: Several studies have shown a relation between obesity and cognitive decline, highlighting a significant global health challenge. In recent years, artificial intelligence (AI) and machine learning (ML) have been integrated into clinical practice for analyzing datasets to identify new risk factors, build predictive models, and develop personalized interventions, thereby providing useful information to healthcare professionals. This systematic review aims to evaluate the potential of AI and ML techniques in addressing the relationship between obesity, its associated health consequences, and cognitive decline. Methods: Systematic searches were performed in PubMed, Cochrane, Web of Science, Scopus, Embase, and PsycInfo databases, which yielded eight studies. After reading the full text of the selected studies and applying predefined inclusion criteria, eight studies were included based on pertinence and relevance to the topic. Results: The findings underscore the utility of AI and ML in assessing risk and predicting cognitive decline in obese patients. Furthermore, these new technology models identified key risk factors and predictive biomarkers, paving the way for tailored prevention strategies and treatment plans. Conclusions: The early detection, prevention, and personalized interventions facilitated by these technologies can significantly reduce costs and time. Future research should assess ethical considerations, data privacy, and equitable access for all.
Collapse
Affiliation(s)
- Isabella Veneziani
- Department of Nervous System and Behavioural Sciences, Psychology Section, University of Pavia, Piazza Botta, 11, 27100 Pavia, Italy (A.G.)
| | - Alessandro Grimaldi
- Department of Nervous System and Behavioural Sciences, Psychology Section, University of Pavia, Piazza Botta, 11, 27100 Pavia, Italy (A.G.)
| | - Angela Marra
- IRCCS Centro Neurolesi “Bonino-Pulejo”, S.S. 113 Via Palermo C. da Casazza, 98124 Messina, Italy; (A.M.); (E.M.); (S.M.); (A.Q.); (G.M.)
| | - Elisabetta Morini
- IRCCS Centro Neurolesi “Bonino-Pulejo”, S.S. 113 Via Palermo C. da Casazza, 98124 Messina, Italy; (A.M.); (E.M.); (S.M.); (A.Q.); (G.M.)
| | - Laura Culicetto
- IRCCS Centro Neurolesi “Bonino-Pulejo”, S.S. 113 Via Palermo C. da Casazza, 98124 Messina, Italy; (A.M.); (E.M.); (S.M.); (A.Q.); (G.M.)
| | - Silvia Marino
- IRCCS Centro Neurolesi “Bonino-Pulejo”, S.S. 113 Via Palermo C. da Casazza, 98124 Messina, Italy; (A.M.); (E.M.); (S.M.); (A.Q.); (G.M.)
| | - Angelo Quartarone
- IRCCS Centro Neurolesi “Bonino-Pulejo”, S.S. 113 Via Palermo C. da Casazza, 98124 Messina, Italy; (A.M.); (E.M.); (S.M.); (A.Q.); (G.M.)
| | - Giuseppa Maresca
- IRCCS Centro Neurolesi “Bonino-Pulejo”, S.S. 113 Via Palermo C. da Casazza, 98124 Messina, Italy; (A.M.); (E.M.); (S.M.); (A.Q.); (G.M.)
| |
Collapse
|
29
|
Huang HYR, Badar S, Said M, Shah S, Bharadwaj HR, Ramamoorthy K, Alrawashdeh MM, Haroon F, Basit J, Saeed S, Aji N, Tse G, Roy P, Bardhan M. The advent of RNA-based therapeutics for metabolic syndrome and associated conditions: a comprehensive review of the literature. Mol Biol Rep 2024; 51:493. [PMID: 38580818 DOI: 10.1007/s11033-024-09457-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/18/2024] [Indexed: 04/07/2024]
Abstract
Metabolic syndrome (MetS) is a prevalent and intricate health condition affecting a significant global population, characterized by a cluster of metabolic and hormonal disorders disrupting lipid and glucose metabolism pathways. Clinical manifestations encompass obesity, dyslipidemia, insulin resistance, and hypertension, contributing to heightened risks of diabetes and cardiovascular diseases. Existing medications often fall short in addressing the syndrome's multifaceted nature, leading to suboptimal treatment outcomes and potential long-term health risks. This scenario underscores the pressing need for innovative therapeutic approaches in MetS management. RNA-based treatments, employing small interfering RNAs (siRNAs), microRNAs (miRNAs), and antisense oligonucleotides (ASOs), emerge as promising strategies to target underlying biological abnormalities. However, a summary of research available on the role of RNA-based therapeutics in MetS and related co-morbidities is limited. Murine models and human studies have been separately interrogated to determine whether there have been recent advancements in RNA-based therapeutics to offer a comprehensive understanding of treatment available for MetS. In a narrative fashion, we searched for relevant articles pertaining to MetS co-morbidities such as cardiovascular disease, fatty liver disease, dementia, colorectal cancer, and endocrine abnormalities. We emphasize the urgency of exploring novel therapeutic avenues to address the intricate pathophysiology of MetS and underscore the potential of RNA-based treatments, coupled with advanced delivery systems, as a transformative approach for achieving more comprehensive and efficacious outcomes in MetS patients.
Collapse
Affiliation(s)
- Helen Ye Rim Huang
- Faculty of Medicine and Health Science, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Sarah Badar
- Department of Biomedical Science, The University of the West Scotland, Paisley, Scotland
| | - Mohammad Said
- Faculty of Medicine and Health Science, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Siddiqah Shah
- Faculty of Medicine and Health Science, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Krishna Ramamoorthy
- Department of Biochemistry and Microbiology, Rutgers University-New Brunswick, Brunswick, NJ, USA
| | | | | | - Jawad Basit
- Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Sajeel Saeed
- Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Narjiss Aji
- Faculty of Medicine and Health, McGill University, Montreal, QC, Canada
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
- School of Nursing and Health Studies, Hong Kong Metropolitan University, Hong Kong, China
| | - Priyanka Roy
- Directorate of Factories, Department of Labour, Government of West Bengal, Kolkata, India
| | - Mainak Bardhan
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA.
| |
Collapse
|
30
|
McGill CJ, Christensen A, Qian W, Thorwald MA, Lugo JG, Namvari S, White OS, Finch CE, Benayoun BA, Pike CJ. Protection against APOE4 -associated aging phenotypes with the longevity-promoting intervention 17α-estradiol in male mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584678. [PMID: 38559059 PMCID: PMC10980056 DOI: 10.1101/2024.03.12.584678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The apolipoprotein ε4 allele ( APOE4 ) is associated with decreased longevity, increased vulnerability to age-related declines, and disorders across multiple systems. Interventions that promote healthspan and lifespan represent a promising strategy to attenuate the development of APOE4 -associated aging phenotypes. Here we studied the ability of the longevity-promoting intervention 17α-estradiol (17αE2) to protect against age-related impairments in APOE4 versus the predominant APOE3 genotype using early middle-aged mice with knock-in of human APOE alleles. Beginning at age 10 months, male APOE3 or APOE4 mice were treated for 20 weeks with 17αE2 or vehicle then compared for indices of aging phenotypes body-wide. Across peripheral and neural measures, APOE4 was associated with poorer outcomes. Notably, 17αE2 treatment improved outcomes in a genotype-dependent manner favoring APOE4 mice. These data demonstrate a positive APOE4 bias in 17αE2-mediated healthspan actions, suggesting that longevity-promoting interventions may be useful in mitigating deleterious age-related risks associated with APOE4 genotype.
Collapse
|
31
|
Zhang J, Pandey M, Awe A, Lue N, Kittock C, Fikse E, Degner K, Staples J, Mokhasi N, Chen W, Yang Y, Adikaram P, Jacob N, Greenfest-Allen E, Thomas R, Bomeny L, Zhang Y, Petros TJ, Wang X, Li Y, Simonds WF. The association of GNB5 with Alzheimer disease revealed by genomic analysis restricted to variants impacting gene function. Am J Hum Genet 2024; 111:473-486. [PMID: 38354736 PMCID: PMC10940018 DOI: 10.1016/j.ajhg.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
Disease-associated variants identified from genome-wide association studies (GWASs) frequently map to non-coding areas of the genome such as introns and intergenic regions. An exclusive reliance on gene-agnostic methods of genomic investigation could limit the identification of relevant genes associated with polygenic diseases such as Alzheimer disease (AD). To overcome such potential restriction, we developed a gene-constrained analytical method that considers only moderate- and high-risk variants that affect gene coding sequences. We report here the application of this approach to publicly available datasets containing 181,388 individuals without and with AD and the resulting identification of 660 genes potentially linked to the higher AD prevalence among Africans/African Americans. By integration with transcriptome analysis of 23 brain regions from 2,728 AD case-control samples, we concentrated on nine genes that potentially enhance the risk of AD: AACS, GNB5, GNS, HIPK3, MED13, SHC2, SLC22A5, VPS35, and ZNF398. GNB5, the fifth member of the heterotrimeric G protein beta family encoding Gβ5, is primarily expressed in neurons and is essential for normal neuronal development in mouse brain. Homozygous or compound heterozygous loss of function of GNB5 in humans has previously been associated with a syndrome of developmental delay, cognitive impairment, and cardiac arrhythmia. In validation experiments, we confirmed that Gnb5 heterozygosity enhanced the formation of both amyloid plaques and neurofibrillary tangles in the brains of AD model mice. These results suggest that gene-constrained analysis can complement the power of GWASs in the identification of AD-associated genes and may be more broadly applicable to other polygenic diseases.
Collapse
Affiliation(s)
- Jianhua Zhang
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Mritunjay Pandey
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Adam Awe
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicole Lue
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Claire Kittock
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Emma Fikse
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Katherine Degner
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jenna Staples
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Neha Mokhasi
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Weiping Chen
- Genomic Core, National Institute of Diabetes and Digestive and Kidney Diseases, Bldg. 8/Rm 1A11, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yanqin Yang
- Laboratory of Transplantation Genomics, National Heart Lung and Blood Institute, Bldg. 10/Rm 7S261, National Institutes of Health, Bethesda, MD 20892, USA
| | - Poorni Adikaram
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nirmal Jacob
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Emily Greenfest-Allen
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rachel Thomas
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Laura Bomeny
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yajun Zhang
- Unit on Cellular and Molecular Neurodevelopment, Bldg. 35/Rm 3B 1002, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Timothy J Petros
- Unit on Cellular and Molecular Neurodevelopment, Bldg. 35/Rm 3B 1002, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xiaowen Wang
- Partek Incorporated, 12747 Olive Boulevard, St. Louis, MO 63141, USA
| | - Yulong Li
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - William F Simonds
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
32
|
Dorranipour D, Pourjafari F, Malekpour-Afshar R, Basiri M, Hosseini M. Astrocyte response to melatonin treatment in rats under high-carbohydrate high-fat diet. J Chem Neuroanat 2024; 136:102389. [PMID: 38215799 DOI: 10.1016/j.jchemneu.2024.102389] [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: 10/13/2023] [Revised: 12/29/2023] [Accepted: 01/07/2024] [Indexed: 01/14/2024]
Abstract
The involvement of consumption of high-carbohydrate high-fat (HCHF) diet in cognitive impairment is attributed, at least in part, to the activation of astrocytes, which contributes to the development of neuroinflammation, oxidative stress, and subsequent cognitive deficits. This study aimed to assess the influence of melatonin on cognitive impairment and astrogliosis induced by the HCHF diet in rats. Male Wistar rats were fed an HCHF diet for eight weeks to induce obesity and metabolic syndrome. Subsequently, they received oral melatonin treatment for four weeks at doses of 5 mg/kg, 10 mg/kg, and 30 mg/kg, alongside the HCHF diet. Cognitive function was evaluated using the Y-maze test, while the levels of proinflammatory cytokines, oxidative stress, and the number glial fibrillary acidic protein (GFAP) positive cells were assessed in the hippocampi and hypothalamus. The consumption of the HCHF diet resulted in weight gain, hyperlipidemia, impaired glucose tolerance, cognitive decline, neuroinflammation, oxidative stress damage, and astrogliosis in rats. Although melatonin treatment did not demonstrate beneficial effects on blood glucose and lipid metabolism, it improved the impaired working memory caused by the HCHF diet. Melatonin exhibited a dose-dependent reduction of astrogliosis, neuroinflammation, and lipid peroxidation while restored superoxide dismutase in the hippocampus and hypothalamus of HCHF diet-treated rats. These findings provide evidence that melatonin inhibits astrocyte activation, thereby attenuating inflammation and minimizing oxidative stress damage induced by the HCHF diet.
Collapse
Affiliation(s)
- Davood Dorranipour
- Department of Anatomical Sciences, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Fahimeh Pourjafari
- Department of Anatomical Sciences, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Reza Malekpour-Afshar
- Pathology and Stem Cells Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohsen Basiri
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mehran Hosseini
- Department of Anatomical Sciences, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
33
|
Mäkinen EE, Lensu S, Wikgren J, Pekkala S, Koch LG, Britton SL, Nokia MS. Intrinsic running capacity associates with hippocampal electrophysiology and long-term potentiation in rats. Neurosci Lett 2024; 823:137665. [PMID: 38301912 DOI: 10.1016/j.neulet.2024.137665] [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: 09/05/2023] [Revised: 01/11/2024] [Accepted: 01/29/2024] [Indexed: 02/03/2024]
Abstract
Good aerobic and metabolic fitness associates with better cognitive performance and brain health. Conversely, poor metabolic health predisposes to neurodegenerative diseases. Our previous findings indicate that rats selectively bred for Low Capacity for Running (LCR) show less synaptic plasticity and more inflammation in the hippocampus and perform worse in tasks requiring flexible cognition than rats bred for High Capacity for Running (HCR). Here we aimed to determine whether hippocampal electrophysiological activity related to learning and memory would be impaired in LCR compared to HCR rats. We also studied whether an exercise intervention could even out the possible differences. We conducted in vivo recordings from the dorsal hippocampus under terminal urethane anesthesia in middle-aged sedentary males and female rats, and in females allowed to access running wheels for 6 weeks. Our results indicate stronger long-term potentiation (LTP) in the CA3-CA1 synapse in HCR than LCR rats, and in female than male rats. Compared to LCR rats, HCR rats had more dentate spikes and more gamma epochs, the occurrence of which also correlated positively with the magnitude of LTP. Voluntary running reduced the differences between female LCR and HCR rats. In conclusion, low innate fitness links to reduced hippocampal function and plasticity which can seems to improve with voluntary aerobic exercise even in middle age.
Collapse
Affiliation(s)
- Elina E Mäkinen
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland.
| | - Sanna Lensu
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland; Centre for Interdisciplinary Brain Research, Department of Psychology, University of Jyväskylä, Jyväskylä, Finland
| | - Jan Wikgren
- Centre for Interdisciplinary Brain Research, Department of Psychology, University of Jyväskylä, Jyväskylä, Finland
| | - Satu Pekkala
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Lauren G Koch
- Department of Physiology and Pharmacology, The University of Toledo, Toledo, OH, USA
| | - Steven L Britton
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Miriam S Nokia
- Centre for Interdisciplinary Brain Research, Department of Psychology, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
34
|
Villablanca A, Dugger BN, Nuthikattu S, Chauhan J, Cheung S, Chuah CN, Garrison SL, Milenkovic D, Norman JE, Oliveira LC, Smith BP, Brown SD. How cy pres promotes transdisciplinary convergence science: an academic health center for women's cardiovascular and brain health. J Clin Transl Sci 2024; 8:e16. [PMID: 38384925 PMCID: PMC10880003 DOI: 10.1017/cts.2023.705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 11/15/2023] [Accepted: 12/15/2023] [Indexed: 02/23/2024] Open
Abstract
Cardiovascular disease (CVD) is largely preventable, and the leading cause of death for men and women. Though women have increased life expectancy compared to men, there are marked sex disparities in prevalence and risk of CVD-associated mortality and dementia. Yet, the basis for these and female-male differences is not completely understood. It is increasingly recognized that heart and brain health represent a lifetime of exposures to shared risk factors (including obesity, hyperlipidemia, diabetes, and hypertension) that compromise cerebrovascular health. We describe the process and resources for establishing a new research Center for Women's Cardiovascular and Brain Health at the University of California, Davis as a model for: (1) use of the cy pres principle for funding science to improve health; (2) transdisciplinary collaboration to leapfrog progress in a convergence science approach that acknowledges and addresses social determinants of health; and (3) training the next generation of diverse researchers. This may serve as a blueprint for future Centers in academic health institutions, as the cy pres mechanism for funding research is a unique mechanism to leverage residual legal settlement funds to catalyze the pace of scientific discovery, maximize innovation, and promote health equity in addressing society's most vexing health problems.
Collapse
Affiliation(s)
- Amparo Villablanca
- Department of Internal Medicine, University of California, Davis, CA, USA
| | - Brittany N. Dugger
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA, USA
| | | | - Joohi Chauhan
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA, USA
- Department of Computer Engineering, University of California, Davis, CA, USA
| | - Samson Cheung
- Department of Computer Engineering, University of California, Davis, CA, USA
| | - Chen-Nee Chuah
- Department of Computer Engineering, University of California, Davis, CA, USA
| | - Siedah L. Garrison
- Department of Internal Medicine, University of California, Davis, CA, USA
| | | | - Jennifer E. Norman
- Department of Internal Medicine, University of California, Davis, CA, USA
| | - Luca Cerny Oliveira
- Department of Computer Engineering, University of California, Davis, CA, USA
| | - Bridgette P. Smith
- Department of Internal Medicine, University of California, Davis, CA, USA
| | - Susan D. Brown
- Department of Internal Medicine, University of California, Davis, CA, USA
| |
Collapse
|
35
|
Frentz I, Marcolini S, Schneider CCI, Ikram MA, Mondragon J, De Deyn PP. Metabolic Syndrome Status Changes and Cognitive Functioning: Insights from the Lifelines Cohort Study. J Prev Alzheimers Dis 2024; 11:1283-1290. [PMID: 39350374 PMCID: PMC11436447 DOI: 10.14283/jpad.2024.90] [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: 02/05/2024] [Accepted: 03/13/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Metabolic syndrome is associated with increased risk of dementia. Yet, findings on how longitudinal development of metabolic syndrome status affects cognition remain controversial. OBJECTIVES This study examines whether individuals with different changes in metabolic syndrome status differ in cognitive functioning. Additionally, the prevalence of metabolic syndrome within the Lifelines population-based study is investigated. DESIGN 14609 Lifelines participants (mean age 60.8, 56.4% women) were divided into four groups based on their metabolic syndrome status changes between 2007-2013 (1) and between 2014-2017 (2): without metabolic syndrome (N=10863; absent at 1 and 2), de novo metabolic syndrome (N=1340; absent at 1 and present at 2), remitting metabolic syndrome (N=825; present at 1 and absent at 2), and persistent metabolic syndrome (N=1581; present at 1 and 2). ANCOVA models were employed to assess group differences in psychomotor function, visual attention, visual learning, and working memory assessed using the Cogstate Brief Battery. RESULTS Accounting for education, age, sex, and time between examinations, groups did not statistically differ in any of the four cognitive outcomes. The prevalence of metabolic syndrome within the Lifelines population increased with age and differed among men and women. CONCLUSION Performance in psychomotor function, visual attention, visual learning, and working memory measured by the Cogstate Brief Battery did not differ between individuals with different changes in metabolic syndrome. The length of metabolic syndrome exposure was unknown, making our results exploratory and calling for future studies addressing this gap.
Collapse
Affiliation(s)
- I Frentz
- Sofia Marcolini, Department of Neurology, University Medical Center Groningen, Groningen, the Netherlands Hanzeplein 1, , 0503612603
| | | | | | | | | | | |
Collapse
|
36
|
Patwekar M, Patwekar F, Khan S, Sharma R, Kumar D. Navigating the Alzheimer's Treatment Landscape: Unraveling Amyloid-beta Complexities and Pioneering Precision Medicine Approaches. Curr Top Med Chem 2024; 24:1665-1682. [PMID: 38644708 DOI: 10.2174/0115680266295495240415114919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/26/2024] [Accepted: 03/05/2024] [Indexed: 04/23/2024]
Abstract
A variety of cutting-edge methods and good knowledge of the illness's complex causes are causing a sea change in the field of Alzheimer's Disease (A.D.) research and treatment. Precision medicine is at the vanguard of this change, where individualized treatment plans based on genetic and biomarker profiles give a ray of hope for customized therapeutics. Combination therapies are becoming increasingly popular as a way to address the multifaceted pathology of Alzheimer's by simultaneously attacking Aβ plaques, tau tangles, neuroinflammation, and other factors. The article covers several therapeutic design efforts, including BACE inhibitors, gamma- secretase modulators, monoclonal antibodies (e.g., Aducanumab and Lecanemab), and anti- Aβ vaccinations. While these techniques appear promising, clinical development faces safety concerns and uneven efficacy. To address the complicated Aβ pathology in Alzheimer's disease, a multimodal approach is necessary. The statement emphasizes the continued importance of clinical trials in addressing safety and efficacy concerns. Looking ahead, it suggests that future treatments may take into account genetic and biomarker traits in order to provide more personalized care. Therapies targeting Aβ, tau tangles, neuroinflammation, and novel drug delivery modalities are planned. Nanoparticles and gene therapies are only two examples of novel drug delivery methods that have the potential to deliver treatments more effectively, with fewer side effects, and with better therapeutic results. In addition, medicines that target tau proteins in addition to Aβ are in the works. Early intervention, based on precise biomarkers, is a linchpin of Alzheimer's care, emphasizing the critical need for detecting the disease at its earliest stages. Lifestyle interventions, encompassing diet, exercise, cognitive training, and social engagement, are emerging as key components in the fight against cognitive decline. Data analytics and art are gaining prominence as strategies to mitigate the brain's inflammatory responses. To pool knowledge and resources in the fight against Alzheimer's, international cooperation between scientists, doctors, and pharmaceutical companies is still essential. In essence, a complex, individualized, and collaborative strategy will characterize Alzheimer's research and therapy in the future. Despite obstacles, these encouraging possibilities show the ongoing commitment of the scientific and medical communities to combat A.D. head-on, providing a glimmer of hope to the countless people and families touched by this savage sickness.
Collapse
Affiliation(s)
- Mohsina Patwekar
- Department of Pharmacology, Luqman College of Pharmacy, P.B. 86, old Jewargi road, Gulbarga, Karnataka, 585102, India
| | - Faheem Patwekar
- Department of Pharmacognosy, Luqman College of Pharmacy, P.B. 86, old Jewargi Road, Gulbarga, Karnataka, 585102, India
| | - Shahzad Khan
- Department of Biomedical Sciences, College of Clinical Pharmacy, King Faisal University, Al Ahsa City, Saudi Arabia
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Dileep Kumar
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra 411038, India
- UC Davis Comprehensive Cancer Center, University of California, Davis, One Shields Ave, Davis, CA 95616, USA
| |
Collapse
|
37
|
Sachdeva P, Narayanan KB, Sinha JK, Gupta S, Ghosh S, Singh KK, Bhaskar R, Almutary AG, Zothantluanga JH, Kotta KK, Nelson VK, Paiva-Santos AC, Abomughaid MM, Kamal M, Iqbal D, ALHarbi MH, ALMutairi AA, Dewanjee S, Nuli MV, Vippamakula S, Jha SK, Ojha S, Jha NK. Recent Advances in Drug Delivery Systems Targeting Insulin Signalling for the Treatment of Alzheimer's Disease. J Alzheimers Dis 2024; 98:1169-1179. [PMID: 38607755 DOI: 10.3233/jad-231181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized by the accumulation of neurofibrillary tangles and amyloid-β plaques. Recent research has unveiled the pivotal role of insulin signaling dysfunction in the pathogenesis of AD. Insulin, once thought to be unrelated to brain function, has emerged as a crucial factor in neuronal survival, synaptic plasticity, and cognitive processes. Insulin and the downstream insulin signaling molecules are found mainly in the hippocampus and cortex. Some molecules responsible for dysfunction in insulin signaling are GSK-3β, Akt, PI3K, and IRS. Irregularities in insulin signaling or insulin resistance may arise from changes in the phosphorylation levels of key molecules, which can be influenced by both stimulation and inactivity. This, in turn, is believed to be a crucial factor contributing to the development of AD, which is characterized by oxidative stress, neuroinflammation, and other pathological hallmarks. Furthermore, this route is known to be indirectly influenced by Nrf2, NF-κB, and the caspases. This mini-review delves into the intricate relationship between insulin signaling and AD, exploring how disruptions in this pathway contribute to disease progression. Moreover, we examine recent advances in drug delivery systems designed to target insulin signaling for AD treatment. From oral insulin delivery to innovative nanoparticle approaches and intranasal administration, these strategies hold promise in mitigating the impact of insulin resistance on AD. This review consolidates current knowledge to shed light on the potential of these interventions as targeted therapeutic options for AD.
Collapse
Affiliation(s)
- Punya Sachdeva
- GloNeuro, Noida, Uttar Pradesh, India
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| | - Kannan Badri Narayanan
- School of Chemical Engineering, Yeungnam University, Gyeongsan, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Republic of Korea
| | | | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology, Rajiv Gandhi InfoTech Park, Hinjawadi, Pune, Maharashtra, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Republic of Korea
| | - Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | - James H Zothantluanga
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, Assam, India
| | - Kranthi Kumar Kotta
- College of Pharmaceutical Sciences, Dayananda Sagar University, Bengaluru, Karnataka, India
| | - Vinod Kumar Nelson
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy of the University of Coimbra, REQUIMTE/LAQV, Group of Pharmaceutical Technology, University of Coimbra, Coimbra, Portugal
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia
| | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah, Saudi Arabia
| | - Mohammed Hamoud ALHarbi
- Department of Infection Control, Senior Consultant of Public Health, King Khalid Hospital, Al Majmaah, Ministry of Health, Saudi Arabia
| | - Awadh Aedh ALMutairi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, Saudi Arabia
| | - Saikat Dewanjee
- Department of Pharmaceutical Technology, Advanced Pharmacognosy Research Laboratory, Jadavpur University, Kolkata, India
| | - Mohana Vamsi Nuli
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India
| | - Shanmugam Vippamakula
- MB School of Pharmaceutical Sciences, Mohan Babu University, A. Rangampet, Tirupati, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, Delhi, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Niraj Kumar Jha
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India
- Department of Biotechnology, School of Applied and Life Sciences (SALS), Uttaranchal University, Dehradun, India
| |
Collapse
|
38
|
Neațu M, Covaliu A, Ioniță I, Jugurt A, Davidescu EI, Popescu BO. Monoclonal Antibody Therapy in Alzheimer's Disease. Pharmaceutics 2023; 16:60. [PMID: 38258071 PMCID: PMC11154277 DOI: 10.3390/pharmaceutics16010060] [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: 11/03/2023] [Revised: 12/22/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
Alzheimer's disease is a neurodegenerative condition marked by the progressive deterioration of cognitive abilities, memory impairment, and the accumulation of abnormal proteins, specifically beta-amyloid plaques and tau tangles, within the brain. Despite extensive research efforts, Alzheimer's disease remains without a cure, presenting a significant global healthcare challenge. Recently, there has been an increased focus on antibody-based treatments as a potentially effective method for dealing with Alzheimer's disease. This paper offers a comprehensive overview of the current status of research on antibody-based molecules as therapies for Alzheimer's disease. We will briefly mention their mechanisms of action, therapeutic efficacy, and safety profiles while addressing the challenges and limitations encountered during their development. We also highlight some crucial considerations in antibody-based treatment development, including patient selection criteria, dosing regimens, or safety concerns. In conclusion, antibody-based therapies present a hopeful outlook for addressing Alzheimer's disease. While challenges remain, the accumulating evidence suggests that these therapies may offer substantial promise in ameliorating or preventing the progression of this debilitating condition, thus potentially enhancing the quality of life for the millions of individuals and families affected by Alzheimer's disease worldwide.
Collapse
Affiliation(s)
- Monica Neațu
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (A.C.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Anca Covaliu
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (A.C.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Iulia Ioniță
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (A.C.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Ana Jugurt
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (A.C.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Eugenia Irene Davidescu
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (A.C.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Bogdan Ovidiu Popescu
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (A.C.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
- Department of Cell Biology, Neurosciences and Experimental Myology, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
39
|
Tang C, Ma Y, Lei X, Ding Y, Yang S, He D. Hypertension linked to Alzheimer's disease via stroke: Mendelian randomization. Sci Rep 2023; 13:21606. [PMID: 38062190 PMCID: PMC10703897 DOI: 10.1038/s41598-023-49087-0] [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: 07/10/2023] [Accepted: 12/04/2023] [Indexed: 12/18/2023] Open
Abstract
This study aimed to investigate the relationship between hypertension and Alzheimer's disease (AD) and demonstrate the key role of stroke in this relationship using mediating Mendelian randomization. AD, a neurodegenerative disease characterized by memory loss, cognitive impairment, and behavioral abnormalities, severely affects the quality of life of patients. Hypertension is an important risk factor for AD. However, the precise mechanism underlying this relationship is unclear. To investigate the relationship between hypertension and AD, we used a mediated Mendelian randomization method and screened for mediating variables between hypertension and AD by setting instrumental variables. The results of the mediated analysis showed that stroke, as a mediating variable, plays an important role in the causal relationship between hypertension and AD. Specifically, the mediated indirect effect value for stroke obtained using multivariate mediated MR analysis was 54.9%. This implies that approximately 55% of the risk of AD owing to hypertension can be attributed to stroke. The results suggest that the increased risk of AD owing to hypertension is mediated through stroke. The finding not only sheds light on the relationship between hypertension and AD but also indicates novel methods for the prevention and treatment of AD. By identifying the critical role of stroke in the link between hypertension and AD, this study provides insights into potential interventions that could mitigate the impact of hypertension on AD. This could help develop personalized treatments and help improve the quality of life of patients with AD who suffer from hypertension.
Collapse
Affiliation(s)
- Chao Tang
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou Province, China
| | - Yayu Ma
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou Province, China
| | - Xiaoyang Lei
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou Province, China
| | - Yaqi Ding
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou Province, China
| | - Sushuang Yang
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou Province, China
| | - Dian He
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou Province, China.
| |
Collapse
|
40
|
Kothawade SM, Buttar HS, Tuli HS, Kaur G. Therapeutic potential of flavonoids in the management of obesity-induced Alzheimer's disease: an overview of preclinical and clinical studies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2813-2830. [PMID: 37231172 DOI: 10.1007/s00210-023-02529-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023]
Abstract
Obesity is a global epidemic that affects people of all ages, genders, and backgrounds. It can lead to a plethora of disorders, including diabetes mellitus, renal dysfunction, musculoskeletal problems, metabolic syndrome, cardiovascular, and neurodegenerative abnormalities. Obesity has also been linked to neurological diseases such as cognitive decline, dementia, and Alzheimer's disease (AD), caused by oxidative stress, pro-inflammatory cytokines, and the production of reactive oxygen free radicals (ROS). Secretion of insulin hormone is impaired in obese people, leading to hyperglycaemia and increased accumulation of amyloid-β in the brain. Acetylcholine, a key neurotransmitter necessary for forming new neuronal connections in the brain, decreases in AD patients. To alleviate acetylcholine deficiency, researchers have proposed dietary interventions and adjuvant therapies that enhance the production of acetylcholine and assist in the management of AD patients. Such measures include dietary intervention with antioxidant and anti-inflammatory flavonoid-rich diets, which have been found to bind to tau receptors, reduce gliosis, and reduce neuroinflammatory markers in animal models. Furthermore, flavonoids like curcumin, resveratrol, epigallocatechin-3-gallate, morin, delphinidins, quercetin, luteolin, and oleocanthal have shown to cause significant reductions in interleukin-1β, increase BDNF levels, stimulate hippocampal neurogenesis and synapse formation, and ultimately prevent the loss of neurons in the brain. Thus, flavonoid-rich nutraceuticals can be a potential cost-effective therapeutic option for treating obesity-induced AD, but further well-designed, randomized, and placebo-controlled clinical studies are needed to assess their optimal dosages, efficacy, and long-term safety of flavonoids in humans. The main objectives of this review are to underscore the therapeutic potential of different nutraceuticals containing flavonoids that can be added in the daily diet of AD patients to enhance acetylcholine and reduce neuronal inflammation in the brain.
Collapse
Affiliation(s)
- Sakshi M Kothawade
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, Mumbai-56, Maharashtra, India
| | - Harpal Singh Buttar
- Department of Pathology & Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Haryana, Mullana, India
| | - Ginpreet Kaur
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, Mumbai-56, Maharashtra, India.
| |
Collapse
|
41
|
Teaney NA, Cyr NE. FoxO1 as a tissue-specific therapeutic target for type 2 diabetes. Front Endocrinol (Lausanne) 2023; 14:1286838. [PMID: 37941908 PMCID: PMC10629996 DOI: 10.3389/fendo.2023.1286838] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/06/2023] [Indexed: 11/10/2023] Open
Abstract
Forkhead box O (FoxO) proteins are transcription factors that mediate many aspects of physiology and thus have been targeted as therapeutics for several diseases including metabolic disorders such as type 2 diabetes mellitus (T2D). The role of FoxO1 in metabolism has been well studied, but recently FoxO1's potential for diabetes prevention and therapy has been debated. For example, studies have shown that increased FoxO1 activity in certain tissue types contributes to T2D pathology, symptoms, and comorbidities, yet in other tissue types elevated FoxO1 has been reported to alleviate symptoms associated with diabetes. Furthermore, studies have reported opposite effects of active FoxO1 in the same tissue type. For example, in the liver, FoxO1 contributes to T2D by increasing hepatic glucose production. However, FoxO1 has been shown to either increase or decrease hepatic lipogenesis as well as adipogenesis in white adipose tissue. In skeletal muscle, FoxO1 reduces glucose uptake and oxidation, promotes lipid uptake and oxidation, and increases muscle atrophy. While many studies show that FoxO1 lowers pancreatic insulin production and secretion, others show the opposite, especially in response to oxidative stress and inflammation. Elevated FoxO1 in the hypothalamus increases the risk of developing T2D. However, increased FoxO1 may mitigate Alzheimer's disease, a neurodegenerative disease strongly associated with T2D. Conversely, accumulating evidence implicates increased FoxO1 with Parkinson's disease pathogenesis. Here we review FoxO1's actions in T2D conditions in metabolic tissues that abundantly express FoxO1 and highlight some of the current studies targeting FoxO1 for T2D treatment.
Collapse
Affiliation(s)
- Nicole A. Teaney
- Stonehill College, Neuroscience Program, Easton, MA, United States
| | - Nicole E. Cyr
- Stonehill College, Neuroscience Program, Easton, MA, United States
- Stonehill College, Department of Biology, Easton, MA, United States
| |
Collapse
|
42
|
Masliukov PM. Changes of Signaling Pathways in Hypothalamic Neurons with Aging. Curr Issues Mol Biol 2023; 45:8289-8308. [PMID: 37886966 PMCID: PMC10605528 DOI: 10.3390/cimb45100523] [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: 08/28/2023] [Revised: 10/01/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
The hypothalamus is an important regulator of autonomic and endocrine functions also involved in aging regulation. The aging process in the hypothalamus is accompanied by disturbed intracellular signaling including insulin/insulin-like growth factor-1 (IGF-1)/growth hormone (GH), phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (AKT)/the mammalian target of rapamycin (mTOR), mitogen activated protein kinase (MAPK), janus kinase (JAK)/signal transducer and activator of transcription (STAT), AMP-activated protein kinase (AMPK), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-ĸB), and nitric oxide (NO). In the current review, I have summarized the current understanding of the changes in the above-mentioned pathways in aging with a focus on hypothalamic alterations.
Collapse
Affiliation(s)
- Petr M Masliukov
- Department Normal Physiology, Yaroslavl State Medical University, ul. Revoliucionnaya 5, 150000 Yaroslavl, Russia
| |
Collapse
|
43
|
Puranik N, Yadav D, Song M. Advancements in the Application of Nanomedicine in Alzheimer's Disease: A Therapeutic Perspective. Int J Mol Sci 2023; 24:14044. [PMID: 37762346 PMCID: PMC10530821 DOI: 10.3390/ijms241814044] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that affects most people worldwide. AD is a complex central nervous system disorder. Several drugs have been designed to cure AD, but with low success rates. Because the blood-brain and blood-cerebrospinal fluid barriers are two barriers that protect the central nervous system, their presence has severely restricted the efficacy of many treatments that have been studied for AD diagnosis and/or therapy. The use of nanoparticles for the diagnosis and treatment of AD is the focus of an established and rapidly developing field of nanomedicine. Recent developments in nanomedicine have made it possible to effectively transport drugs to the brain. However, numerous obstacles remain to the successful use of nanomedicines in clinical settings for AD treatment. Furthermore, given the rapid advancement in nanomedicine therapeutics, better outcomes for patients with AD can be anticipated. This article provides an overview of recent developments in nanomedicine using different types of nanoparticles for the management and treatment of AD.
Collapse
Affiliation(s)
| | | | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (N.P.); (D.Y.)
| |
Collapse
|
44
|
Szukiewicz D. Molecular Mechanisms for the Vicious Cycle between Insulin Resistance and the Inflammatory Response in Obesity. Int J Mol Sci 2023; 24:9818. [PMID: 37372966 DOI: 10.3390/ijms24129818] [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: 05/12/2023] [Revised: 05/31/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
The comprehensive anabolic effects of insulin throughout the body, in addition to the control of glycemia, include ensuring lipid homeostasis and anti-inflammatory modulation, especially in adipose tissue (AT). The prevalence of obesity, defined as a body mass index (BMI) ≥ 30 kg/m2, has been increasing worldwide on a pandemic scale with accompanying syndemic health problems, including glucose intolerance, insulin resistance (IR), and diabetes. Impaired tissue sensitivity to insulin or IR paradoxically leads to diseases with an inflammatory component despite hyperinsulinemia. Therefore, an excess of visceral AT in obesity initiates chronic low-grade inflammatory conditions that interfere with insulin signaling via insulin receptors (INSRs). Moreover, in response to IR, hyperglycemia itself stimulates a primarily defensive inflammatory response associated with the subsequent release of numerous inflammatory cytokines and a real threat of organ function deterioration. In this review, all components of this vicious cycle are characterized with particular emphasis on the interplay between insulin signaling and both the innate and adaptive immune responses related to obesity. Increased visceral AT accumulation in obesity should be considered the main environmental factor responsible for the disruption in the epigenetic regulatory mechanisms in the immune system, resulting in autoimmunity and inflammation.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
45
|
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
|