1
|
Zhang Y, Liang Y, Gu Y. The dopaminergic system and Alzheimer's disease. Neural Regen Res 2025; 20:2495-2512. [PMID: 39314145 DOI: 10.4103/nrr.nrr-d-24-00230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 07/31/2024] [Indexed: 09/25/2024] Open
Abstract
Alzheimer's disease is a common neurodegenerative disorder in older adults. Despite its prevalence, its pathogenesis remains unclear. In addition to the most widely accepted causes, which include excessive amyloid-beta aggregation, tau hyperphosphorylation, and deficiency of the neurotransmitter acetylcholine, numerous studies have shown that the dopaminergic system is also closely associated with the occurrence and development of this condition. Dopamine is a crucial catecholaminergic neurotransmitter in the human body. Dopamine-associated treatments, such as drugs that target dopamine receptor D and dopamine analogs, can improve cognitive function and alleviate psychiatric symptoms as well as ameliorate other clinical manifestations. However, therapeutics targeting the dopaminergic system are associated with various adverse reactions, such as addiction and exacerbation of cognitive impairment. This review summarizes the role of the dopaminergic system in the pathology of Alzheimer's disease, focusing on currently available dopamine-based therapies for this disorder and the common side effects associated with dopamine-related drugs. The aim of this review is to provide insights into the potential connections between the dopaminergic system and Alzheimer's disease, thus helping to clarify the mechanisms underlying the condition and exploring more effective therapeutic options.
Collapse
Affiliation(s)
- Yuhan Zhang
- International Medical College, Chongqing Medical University, Chongqing, China
| | - Yuan Liang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yixue Gu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| |
Collapse
|
2
|
August I, Gagneux P, Semendeferi K, Marchetto MC. Evolution of Human Susceptibility to Alzheimer's Disease: A Review of Hypotheses and Comparative Evidence. Evol Anthropol 2025; 34:e22054. [PMID: 39806778 DOI: 10.1002/evan.22054] [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: 08/24/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025]
Abstract
Primates rely on memory to navigate both physical and social environments and in humans, loss of memory function leads to devastating consequences. Alzheimer's disease (AD) is a neurodegenerative disease which begins by impacting memory functioning and is ultimately fatal. AD is common across human populations and its prevalence is predicted to rise with increases in the aging population. Despite this, the full AD phenotype has not been observed in any other nonhuman primate species. While a significant amount of research has been devoted to understanding the immediate mechanisms involved in AD pathogenesis in humans, less research has focused on why humans are particularly vulnerable to neurodegenerative diseases like AD. Here we explore hypotheses on the evolution of distinct human susceptibility to AD and place these in the context of findings from comparative neuroanatomical and molecular studies and discuss recent evidence for evolutionary changes protective against AD in the primate lineage.
Collapse
Affiliation(s)
- Isabel August
- Department of Anthropology, University of California San Diego, La Jolla, California, USA
| | - Pascal Gagneux
- Department of Anthropology, University of California San Diego, La Jolla, California, USA
- Department of Pathology, University of California San Diego, La Jolla, California, USA
- Center for Academic Research and Training in Anthropogeny (CARTA), La Jolla, California, USA
| | - Katerina Semendeferi
- Department of Anthropology, University of California San Diego, La Jolla, California, USA
- Center for Academic Research and Training in Anthropogeny (CARTA), La Jolla, California, USA
| | - Maria Carolina Marchetto
- Department of Anthropology, University of California San Diego, La Jolla, California, USA
- Center for Academic Research and Training in Anthropogeny (CARTA), La Jolla, California, USA
| |
Collapse
|
3
|
Cervera-Juanes R, Zimmerman KD, Wilhelm L, Zhu D, Bodie J, Kohama SG, Urbanski HF. Modulation of neural gene networks by estradiol in old rhesus macaque females. GeroScience 2024; 46:5819-5841. [PMID: 38509416 PMCID: PMC11493911 DOI: 10.1007/s11357-024-01133-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
The postmenopausal decrease in circulating estradiol (E2) levels has been shown to contribute to several adverse physiological and psychiatric effects. To elucidate the molecular effects of E2 on the brain, we examined differential gene expression and DNA methylation (DNAm) patterns in the nonhuman primate brain following ovariectomy (Ov) and subsequent subcutaneous bioidentical E2 chronic treatment. We identified several dysregulated molecular networks, including MAPK signaling and dopaminergic synapse response, that are associated with ovariectomy and shared across two different brain areas, the occipital cortex (OC) and prefrontal cortex (PFC). The finding that hypomethylation (p = 1.6 × 10-51) and upregulation (p = 3.8 × 10-3) of UBE2M across both brain regions provide strong evidence for molecular differences in the brain induced by E2 depletion. Additionally, differential expression (p = 1.9 × 10-4; interaction p = 3.5 × 10-2) of LTBR in the PFC provides further support for the role E2 plays in the brain, by demonstrating that the regulation of some genes that are altered by ovariectomy may also be modulated by Ov followed by hormone replacement therapy (HRT). These results present real opportunities to understand the specific biological mechanisms that are altered with depleted E2. Given E2's potential role in cognitive decline and neuroinflammation, our findings could lead to the discovery of novel therapeutics to slow cognitive decline. Together, this work represents a major step toward understanding molecular changes in the brain that are caused by ovariectomy and how E2 treatment may revert or protect against the negative neuro-related consequences caused by a depletion in estrogen as women approach menopause.
Collapse
Affiliation(s)
- Rita Cervera-Juanes
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
- Center for Precision Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
| | - Kip D Zimmerman
- Center for Precision Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Larry Wilhelm
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Dongqin Zhu
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Jessica Bodie
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Steven G Kohama
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
| | - Henryk F Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| |
Collapse
|
4
|
Cacabelos R, Martínez-Iglesias O, Cacabelos N, Carrera I, Corzo L, Naidoo V. Therapeutic Options in Alzheimer's Disease: From Classic Acetylcholinesterase Inhibitors to Multi-Target Drugs with Pleiotropic Activity. Life (Basel) 2024; 14:1555. [PMID: 39768263 PMCID: PMC11678002 DOI: 10.3390/life14121555] [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: 10/24/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
Alzheimer's disease (AD) is a complex/multifactorial brain disorder involving hundreds of defective genes, epigenetic aberrations, cerebrovascular alterations, and environmental risk factors. The onset of the neurodegenerative process is triggered decades before the first symptoms appear, probably due to a combination of genomic and epigenetic phenomena. Therefore, the primary objective of any effective treatment is to intercept the disease process in its presymptomatic phases. Since the approval of acetylcholinesterase inhibitors (Tacrine, Donepezil, Rivastigmine, Galantamine) and Memantine, between 1993 and 2003, no new drug was approved by the FDA until the advent of immunotherapy with Aducanumab in 2021 and Lecanemab in 2023. Over the past decade, more than 10,000 new compounds with potential action on some pathogenic components of AD have been tested. The limitations of these anti-AD treatments have stimulated the search for multi-target (MT) drugs. In recent years, more than 1000 drugs with potential MT function have been studied in AD models. MT drugs aim to address the complex and multifactorial nature of the disease. This approach has the potential to offer more comprehensive benefits than single-target therapies, which may be limited in their effectiveness due to the intricate pathology of AD. A strategy still unexplored is the combination of epigenetic drugs with MT agents. Another option could be biotechnological products with pleiotropic action, among which nosustrophine-like compounds could represent an attractive, although not definitive, example.
Collapse
Affiliation(s)
- Ramón Cacabelos
- EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, 15165 Corunna, Spain; (O.M.-I.); (N.C.); (I.C.); (L.C.); (V.N.)
| | | | | | | | | | | |
Collapse
|
5
|
Lacerda RAV, Desio JAF, Kammers CM, Henkes S, Freitas de Sá M, de Souza EF, da Silva DM, Teixeira Pinheiro Gusmão C, Santos JCCD. Sleep disorders and risk of alzheimer's disease: A two-way road. Ageing Res Rev 2024; 101:102514. [PMID: 39317268 DOI: 10.1016/j.arr.2024.102514] [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: 08/08/2024] [Revised: 09/15/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
Substantial sleep impairment in patients with Alzheimer's disease (AD) is one of the emerging points for continued efforts to better understand the disease. Individuals without cognitive decline, an important marker of the clinical phase of AD, may show early alterations in the sleep-wake cycle. The objective of this critical narrative review is to explore the bidirectional pathophysiological correlation between sleep disturbances and Alzheimer's Disease. Specifically, it examines how the disruption of sleep homeostasis in individuals without dementia could contribute to the pathogenesis of AD, and conversely, how neurodegeneration in individuals with Alzheimer's Disease might lead to dysregulation of the sleep-wake cycle. Recent scientific results indicate that sleep disturbances, particularly those related to impaired glymphatic clearance, may act as an important mechanism associated with the genesis of Alzheimer's Disease. Additionally, amyloid deposition and tau protein hyperphosphorylation, along with astrocytic hyperactivation, appear to trigger changes in neurotransmission dynamics in areas related to sleep, which may explain the onset of sleep disturbances in individuals with AD. Disruption of sleep homeostasis appears to be a modifiable risk factor in Alzheimer's disease. Whenever possible, the use of non-pharmacological strategies becomes important in this context. From a different perspective, additional research is needed to understand and treat the dysfunction of the sleep-wake cycle in individuals already affected by AD. Early recognition and correction of sleep disturbances in this population could potentially mitigate the progression of dementia and improve the quality of life for those with AD.
Collapse
Affiliation(s)
| | | | | | - Silvana Henkes
- Lutheran University of Brazil - ULBRA, Carazinho, RS, Brazil
| | | | | | | | | | - Júlio César Claudino Dos Santos
- Medical School of the Christus University Center - UNICHRISTUS, Fortaleza, CE, Brazil; Post-Graduate Program of Morphofunctional Sciences, Federal University of Ceara, Fortaleza, CE, Brazil; Unifacvest University Center - UNIFACVEST, Lages, SC, Brazil.
| |
Collapse
|
6
|
Lee DH, Park EG, Kim JM, Shin HJ, Lee YJ, Jeong HS, Roh HY, Kim WR, Ha H, Kim SW, Choi YH, Kim HS. Genomic analyses of intricate interaction of TE-lncRNA overlapping genes with miRNAs in human diseases. Genes Genomics 2024; 46:1313-1325. [PMID: 39215947 DOI: 10.1007/s13258-024-01547-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/09/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Transposable elements (TEs) are known to be inserted into genome to create transcript isoforms or to generate long non-coding RNA (lncRNA) sequences. The insertion of TEs generates a gene protein sequence within the genome, but also provides a microRNA (miRNA) regulatory region. OBJECTIVE To determine the effect of gene sequence changes caused by TE insertion on miRNA binding and to investigate the formation of an overlapping lncRNA that represses it. METHODS The distribution of overlapping regions between exons and TE regions with lncRNA was examined using the Bedtools. miRNAs that can bind to those overlapping regions were identified through the miRDB web program. For TE-lncRNA overlapping genes, bioinformatic analysis was conducted using DAVID web database. Differential expression analysis was conducted using data from the GEO dataset and TCGA. RESULTS Most TEs were distributed more frequently in untranslated regions than open reading frames. There were 30 annotated TE-lncRNA overlapping genes with same strand that could bind to the same miRNA. As a result of identifying the association between these 30 genes and diseases, TGFB2, FCGR2A, DCTN5, and IFI6 were associated with breast cancer, and HMGCS1, FRMD4A, EDNRB, and SNCA were associated with Alzheimer's disease. Analysis of the GEO and TCGA data showed that the relevant expression of miR-891a and miR-28, which bind to the TE overlapping region of DCTN5 and HMGCS1, decreased. CONCLUSION This study indicates that the interaction between TE-lncRNA overlapping genes and miRNAs can affect disease progression.
Collapse
Affiliation(s)
- Du Hyeong Lee
- Department of Integrated Biological Sciences, Pusan National University, Busan, 46241, Republic of Korea
- Institute of Systems Biology, Pusan National University, Busan, 46241, Republic of Korea
| | - Eun Gyung Park
- Department of Integrated Biological Sciences, Pusan National University, Busan, 46241, Republic of Korea
- Institute of Systems Biology, Pusan National University, Busan, 46241, Republic of Korea
| | - Jung-Min Kim
- Department of Integrated Biological Sciences, Pusan National University, Busan, 46241, Republic of Korea
- Institute of Systems Biology, Pusan National University, Busan, 46241, Republic of Korea
| | - Hae Jin Shin
- Department of Integrated Biological Sciences, Pusan National University, Busan, 46241, Republic of Korea
- Institute of Systems Biology, Pusan National University, Busan, 46241, Republic of Korea
| | - Yun Ju Lee
- Department of Integrated Biological Sciences, Pusan National University, Busan, 46241, Republic of Korea
- Institute of Systems Biology, Pusan National University, Busan, 46241, Republic of Korea
| | - Hyeon-Su Jeong
- Department of Integrated Biological Sciences, Pusan National University, Busan, 46241, Republic of Korea
- Institute of Systems Biology, Pusan National University, Busan, 46241, Republic of Korea
| | - Hyun-Young Roh
- Institute of Systems Biology, Pusan National University, Busan, 46241, Republic of Korea
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan, 46241, Republic of Korea
| | - Woo Ryung Kim
- Department of Integrated Biological Sciences, Pusan National University, Busan, 46241, Republic of Korea
- Institute of Systems Biology, Pusan National University, Busan, 46241, Republic of Korea
| | - Hongseok Ha
- Institute of Endemic Disease, Medical Research Center, Seoul National University, Seoul, 03080, Republic of Korea
| | - Sang-Woo Kim
- Department of Integrated Biological Sciences, Pusan National University, Busan, 46241, Republic of Korea
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan, 46241, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Oriental Medicine, Dong-Eui University, Busan, 47227, Republic of Korea
| | - Heui-Soo Kim
- Institute of Systems Biology, Pusan National University, Busan, 46241, Republic of Korea.
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan, 46241, Republic of Korea.
| |
Collapse
|
7
|
Nasr NN, El-Hagrassi AM, Ahmed YR, Hamed MM. GC/MS and LC-ESI-MS Analysis of Conocarpus erectus Leaves Extract via Regulating Amyloid-β-Peptide, Tau Protein, Neurotransmitters, Inflammation and Oxidative Stress against AlCl 3-Induced Alzheimer's Disease in Rats. Chem Biodivers 2024:e202401960. [PMID: 39367808 DOI: 10.1002/cbdv.202401960] [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: 08/09/2024] [Revised: 09/30/2024] [Accepted: 10/05/2024] [Indexed: 10/07/2024]
Abstract
This study investigated the therapeutic effect of Conocarpus erectus leaves methanolic extract against AlCl3 -induced Alzheimer's disease (AD) in rats comparing with Donepezil-hydrochloride as a reference drug. The bioactive compounds of C. erectus leaves were isolated and identified by GC/MS and LC-ESI-MS analysis. Malondialdehyde (MDA), reduced glutathione (GSH), superoxide dismutase (SOD), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), amyloid-β-peptide (Aβ-peptide), tau protein, acetylcholinesterase (AChE), serotonin (5-HT), dopamine (DA) and nor-adrenaline (NE) levels were estimated. The neuromuscular strength, memory behavior and histopathological examination of cerebral cortex region were also conducted. Forty-three compounds were characterized from the non-polar fraction of C. erectus L. leaves extract and nineteen compounds were identified from the defatted extract. AlCl3- induction caused significant elevation of brain oxidative stress, Aβ-peptide, tau protein, IL-6, TNF-α and AChE levels. A significant decrease in 5-HT, ND and DA levels were noticed. Additionally, AlCl3 reduced neuromuscular strength and compromised memory function. Treatment of AlCl3- induced rats with C. erectuse extract ameliorated these selected parameters by variable degrees. In conclusion, C. erectus protects against AlCl3- induced AD in rats through its antioxidant, anti-inflammatory, and antineutron damage. It could be considered as a new nutraceutical agent for attenuating symptoms associated with Alzheimer's disease.
Collapse
Affiliation(s)
- Noha N Nasr
- Department of Therapeutic Chemistry, National Research Centre, Dokki, Giza, Egypt
| | - Ali M El-Hagrassi
- Phytochemistry and Plant Systematic Department, National Research Centre, 33 El-Buhouth St., Dokki, Giza, Egypt
| | - Yomna R Ahmed
- Department of Therapeutic Chemistry, National Research Centre, Dokki, Giza, Egypt
| | - Manal M Hamed
- Department of Therapeutic Chemistry, National Research Centre, Dokki, Giza, Egypt
| |
Collapse
|
8
|
Saggu S, Bai A, Aida M, Rehman H, Pless A, Ware D, Deak F, Jiao K, Wang Q. Monoamine alterations in Alzheimer's disease and their implications in comorbid neuropsychiatric symptoms. GeroScience 2024:10.1007/s11357-024-01359-x. [PMID: 39331291 DOI: 10.1007/s11357-024-01359-x] [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: 05/22/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder characterized by relentless cognitive decline and the emergence of profoundly disruptive neuropsychiatric symptoms. As the disease progresses, it unveils a formidable array of neuropsychiatric manifestations, including debilitating depression, anxiety, agitation, and distressing episodes of psychosis. The intricate web of the monoaminergic system, governed by serotonin, dopamine, and norepinephrine, significantly influences our mood, cognition, and behavior. Emerging evidence suggests that dysregulation and degeneration of this system occur early in AD, leading to notable alterations in these critical neurotransmitters' levels, metabolism, and receptor function. However, how the degeneration of monoaminergic neurons and subsequent compensatory changes contribute to the presentation of neuropsychiatric symptoms observed in Alzheimer's disease remains elusive. This review synthesizes current findings on monoamine alterations in AD and explores how these changes contribute to the neuropsychiatric symptomatology of the disease. By elucidating the biological underpinnings of AD-related psychiatric symptoms, we aim to underscore the complexity and inform innovative approaches for treating neuropsychiatric symptoms in AD.
Collapse
Affiliation(s)
- Shalini Saggu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| | - Ava Bai
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Mae Aida
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Hasibur Rehman
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Andrew Pless
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Destany Ware
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Ferenc Deak
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Kai Jiao
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Qin Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
9
|
Biswas A, Lee S, Cencillo-Abad P, Karmakar M, Patel J, Soudi M, Chanda D. Nanoplasmonic aptasensor for sensitive, selective, and real-time detection of dopamine from unprocessed whole blood. SCIENCE ADVANCES 2024; 10:eadp7460. [PMID: 39231221 PMCID: PMC11373595 DOI: 10.1126/sciadv.adp7460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/29/2024] [Indexed: 09/06/2024]
Abstract
Neurotransmitters are crucial for the proper functioning of neural systems, with dopamine playing a pivotal role in cognition, emotions, and motor control. Dysregulated dopamine levels are linked to various disorders, underscoring the need for accurate detection in research and diagnostics. Single-stranded DNA (ssDNA) aptamers are promising bioreceptors for dopamine detection due to their selectivity, improved stability, and synthesis feasibility. However, discrepancies in dopamine specificity have presented challenges. Here, we surface-functionalized a nano-plasmonic biosensing platform with a dopamine-specific ssDNA aptamer for selective detection. The biosensor, featuring narrowband hybrid plasmonic resonances, achieves high specificity through functionalization with aptamers and passivation processes. Sensitivity and selectivity for dopamine detection are demonstrated across a wide range of concentrations, including in diverse biological samples like protein solutions, cerebrospinal fluid, and whole blood. These results highlight the potential of plasmonic "aptasensors" for developing rapid and accurate diagnostic tools for disease monitoring, medical diagnostics, and targeted therapies.
Collapse
Affiliation(s)
- Aritra Biswas
- CREOL, The College of Optics and Photonics, University of Central Florida, 4304 Scorpius St., Orlando, FL 32816, USA
- Nanoscience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Sang Lee
- Nanoscience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Pablo Cencillo-Abad
- Nanoscience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Manobina Karmakar
- Nanoscience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Jay Patel
- Nanoscience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Mahdi Soudi
- Nanoscience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
- Department of Physics, University of Central Florida, 4111 Libra Drive, Physical Sciences Bldg. 430, Orlando, FL 32816, USA
| | - Debashis Chanda
- CREOL, The College of Optics and Photonics, University of Central Florida, 4304 Scorpius St., Orlando, FL 32816, USA
- Nanoscience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
- Department of Physics, University of Central Florida, 4111 Libra Drive, Physical Sciences Bldg. 430, Orlando, FL 32816, USA
| |
Collapse
|
10
|
Chae S, Lee HJ, Lee HE, Kim J, Jeong YJ, Lin Y, Kim HY, Leriche G, Ehrlich RS, Lingl SC, Seo MD, Lee YH, Yang J, Kim JI, Hoe HS. The dopamine analogue CA140 alleviates AD pathology, neuroinflammation, and rescues synaptic/cognitive functions by modulating DRD1 signaling or directly binding to Abeta. J Neuroinflammation 2024; 21:200. [PMID: 39129007 PMCID: PMC11317008 DOI: 10.1186/s12974-024-03180-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/17/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND We recently reported that the dopamine (DA) analogue CA140 modulates neuroinflammatory responses in lipopolysaccharide-injected wild-type (WT) mice and in 3-month-old 5xFAD mice, a model of Alzheimer's disease (AD). However, the effects of CA140 on Aβ/tau pathology and synaptic/cognitive function and its molecular mechanisms of action are unknown. METHODS To investigate the effects of CA140 on cognitive and synaptic function and AD pathology, 3-month-old WT mice or 8-month-old (aged) 5xFAD mice were injected with vehicle (10% DMSO) or CA140 (30 mg/kg, i.p.) daily for 10, 14, or 17 days. Behavioral tests, ELISA, electrophysiology, RNA sequencing, real-time PCR, Golgi staining, immunofluorescence staining, and western blotting were conducted. RESULTS In aged 5xFAD mice, a model of AD pathology, CA140 treatment significantly reduced Aβ/tau fibrillation, Aβ plaque number, tau hyperphosphorylation, and neuroinflammation by inhibiting NLRP3 activation. In addition, CA140 treatment downregulated the expression of cxcl10, a marker of AD-associated reactive astrocytes (RAs), and c1qa, a marker of the interaction of RAs with disease-associated microglia (DAMs) in 5xFAD mice. CA140 treatment also suppressed the mRNA levels of s100β and cxcl10, markers of AD-associated RAs, in primary astrocytes from 5xFAD mice. In primary microglial cells from 5xFAD mice, CA140 treatment increased the mRNA levels of markers of homeostatic microglia (cx3cr1 and p2ry12) and decreased the mRNA levels of a marker of proliferative region-associated microglia (gpnmb) and a marker of lipid-droplet-accumulating microglia (cln3). Importantly, CA140 treatment rescued scopolamine (SCO)-mediated deficits in long-term memory, dendritic spine number, and LTP impairment. In aged 5xFAD mice, these effects of CA140 treatment on cognitive/synaptic function and AD pathology were regulated by dopamine D1 receptor (DRD1)/Elk1 signaling. In primary hippocampal neurons and WT mice, CA140 treatment promoted long-term memory and dendritic spine formation via effects on DRD1/CaMKIIα and/or ERK signaling. CONCLUSIONS Our results indicate that CA140 improves neuronal/synaptic/cognitive function and ameliorates Aβ/tau pathology and neuroinflammation by modulating DRD1 signaling in primary hippocampal neurons, primary astrocytes/microglia, WT mice, and aged 5xFAD mice.
Collapse
Affiliation(s)
- Sehyun Chae
- Neurovascular Unit, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41068, Republic of Korea
| | - Hyun-Ju Lee
- Neurodegenerative Unit, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41068, Republic of Korea
| | - Ha-Eun Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), 50 UNIST-Gil, Ulsan, 44919, Republic of Korea
| | - Jieun Kim
- Neurodegenerative Unit, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41068, Republic of Korea
| | - Yoo Joo Jeong
- Neurodegenerative Unit, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41068, Republic of Korea
- Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Yuxi Lin
- Biopharmaceutical Research Center, Korea Basic Science Institute (KBSI), Ochang, ChungBuk, 28119, Republic of Korea
| | - Hye Yun Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), 50 UNIST-Gil, Ulsan, 44919, Republic of Korea
| | - Geoffray Leriche
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093-0358, USA
| | - Rachel S Ehrlich
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093-0358, USA
| | - Sascha Castro Lingl
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093-0358, USA
| | - Min-Duk Seo
- College of Pharmacy and Department of Molecular Science and Technology, Ajou University, Suwon, Gyeonggi, 16499, Republic of Korea
| | - Young-Ho Lee
- Biopharmaceutical Research Center, Korea Basic Science Institute (KBSI), Ochang, ChungBuk, 28119, Republic of Korea
- Bio-Analytical Science, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
- Department of Systems Biotechnology, Chung-Ang University, Gyeonggi , 17546, Republic of Korea
| | - Jerry Yang
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093-0358, USA.
| | - Jae-Ick Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), 50 UNIST-Gil, Ulsan, 44919, Republic of Korea.
| | - Hyang-Sook Hoe
- Neurodegenerative Unit, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41068, Republic of Korea.
- Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, 42988, Republic of Korea.
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61 Cheomdan-ro, Dong-gu, Daegu, 41068, Republic of Korea.
| |
Collapse
|
11
|
Zell L, Hofer TS, Schubert M, Popoff A, Höll A, Marschhofer M, Huber-Cantonati P, Temml V, Schuster D. Impact of 2-hydroxypropyl-β-cyclodextrin inclusion complex formation on dopamine receptor-ligand interaction - A case study. Biochem Pharmacol 2024; 226:116340. [PMID: 38848779 DOI: 10.1016/j.bcp.2024.116340] [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: 02/08/2024] [Revised: 05/10/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
The octanol-water distribution coefficient (logP), used as a measure of lipophilicity, plays a major role in the drug design and discovery processes. While average logP values remain unchanged in approved oral drugs since 1983, current medicinal chemistry trends towards increasingly lipophilic compounds that require adapted analytical workflows and drug delivery systems. Solubility enhancers like cyclodextrins (CDs), especially 2-hydroxypropyl-β-CD (2-HP-β-CD), have been studied in vitro and in vivo investigating their ADMET (adsorption, distribution, metabolism, excretion and toxicity)-related properties. However, data is scarce regarding the applicability of CD inclusion complexes (ICs) in vitro compared to pure compounds. In this study, dopamine receptor (DR) ligands were used as a case study, utilizing a combined in silico/in vitro workflow. Media-dependent solubility and IC stoichiometry were investigated using HPLC. NMR was used to observe IC formation-caused chemical shift deviations while in silico approaches utilizing basin hopping global minimization were used to propose putative IC binding modes. A cell-based in vitro homogeneous time-resolved fluorescence (HTRF) assay was used to quantify ligand binding affinity at the DR subtype 2 (D2R). While all ligands showed increased solubility using 2-HP-β-CD, they differed regarding IC stoichiometry and receptor binding affinity. This case study shows that IC-formation was ligand-dependent and sometimes altering in vitro binding. Therefore, IC complex formation can't be recommended as a general means of improving compound solubility for in vitro studies as they may alter ligand binding.
Collapse
Affiliation(s)
- Lukas Zell
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Paracelsus Medical University, 5020 Salzburg, Austria; Research and Innovation Center for Novel Therapies and Regenerative Medicine, Austria
| | - Thomas S Hofer
- Institute of General, Inorganic and Theoretical Chemistry, Center for Biochemistry and Biomedicine, University of Innsbruck, 6020 Innsbruck, Austria
| | - Mario Schubert
- Department of Biosciences and Medical Biology, University of Salzburg, 5020 Salzburg, Austria; Department of Chemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Alexander Popoff
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Paracelsus Medical University, 5020 Salzburg, Austria; Research and Innovation Center for Novel Therapies and Regenerative Medicine, Austria
| | - Anna Höll
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Paracelsus Medical University, 5020 Salzburg, Austria; Research and Innovation Center for Novel Therapies and Regenerative Medicine, Austria
| | - Moritz Marschhofer
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Paracelsus Medical University, 5020 Salzburg, Austria; Research and Innovation Center for Novel Therapies and Regenerative Medicine, Austria
| | - Petra Huber-Cantonati
- Department of Pharmaceutical Biology, Institute of Pharmacy, Paracelsus Medical University, 5020 Salzburg, Austria; Research and Innovation Center for Novel Therapies and Regenerative Medicine, Austria
| | - Veronika Temml
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Paracelsus Medical University, 5020 Salzburg, Austria; Research and Innovation Center for Novel Therapies and Regenerative Medicine, Austria
| | - Daniela Schuster
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Paracelsus Medical University, 5020 Salzburg, Austria; Research and Innovation Center for Novel Therapies and Regenerative Medicine, Austria.
| |
Collapse
|
12
|
Polsinelli AJ, Johnson S, Crouch A, Lane KA, Pena‐Garcia A, Hammers DB, Wang S, Gao S, Apostolova LG. Neuropsychiatric symptom burden in early-onset and late-onset Alzheimer's disease as a function of age. Alzheimers Dement 2024; 20:5481-5491. [PMID: 38958543 PMCID: PMC11350043 DOI: 10.1002/alz.14042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/29/2024] [Accepted: 05/12/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION We examined the burden of neuropsychiatric symptoms (NPSs) in early-onset (EO) and late-onset (LO) Alzheimer's disease (AD) and adjusted for age effects via the inclusion of cognitively unimpaired (CU) individuals. METHODS Cross-sectional data from 2940 EOAD, 8665 LOAD, and 8775 age-stratified CU individuals (early-CU, n = 2433; late-CU, n = 6342) from the National Alzheimer's Coordinating Center database were included. Fisher's exact tests compared EOAD and LOAD on the presence and severity of NPSs. Multiple logistic regression models included an age*diagnosis interaction to examine age effects. RESULTS Presence (ps < 0.0001) and severity (ps < 0.05) of NPS were greater in EOAD than in LOAD. However, after adjusting for base rates in NPS in CU individuals (age effects), only elation and eating behaviors were more frequent in EOAD (ps < 0.05) and nighttime behaviors more frequent and severe in LOAD (ps < 0.05). DISCUSSION Few NPSs were specific to the EOAD versus LOAD. Previous findings of greater NPS burden in EOAD may partially reflect age effects. HIGHLIGHTS Adjusting for age effect, elation and eating problems are more frequent in EOAD. Adjusting for age effect, sleep disturbances are more frequent and severe in LOAD. Age effects underlie higher neuropsychiatric symptom presentation in EOAD than in LOAD.
Collapse
Affiliation(s)
- Angelina J. Polsinelli
- Department of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
- Indiana Alzheimer's Disease Research CenterIndianapolisIndianaUSA
| | - Sierah Johnson
- Department of Psychological SciencesUniversity of IndianapolisIndianapolisIndianaUSA
| | - Adele Crouch
- Science of Nursing Care DepartmentIndiana University School of NursingIndianapolisIndianaUSA
| | - Kathleen A. Lane
- Department of Biostatistics and Health Data ScienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Alex Pena‐Garcia
- College of Osteopathic MedicineMarian UniversityIndianapolisIndianaUSA
| | - Dustin B. Hammers
- Department of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
- Indiana Alzheimer's Disease Research CenterIndianapolisIndianaUSA
| | - Sophia Wang
- Indiana Alzheimer's Disease Research CenterIndianapolisIndianaUSA
- Department of PsychiatryIndiana University School of MedicineIndianapolisIndianaUSA
| | - Sujuan Gao
- Department of Biostatistics and Health Data ScienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Liana G. Apostolova
- Department of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
- Indiana Alzheimer's Disease Research CenterIndianapolisIndianaUSA
| |
Collapse
|
13
|
Giménez S, Millan A, Mora-Morell A, Ayuso N, Gastaldo-Jordán I, Pardo M. Advances in Brain Stimulation, Nanomedicine and the Use of Magnetoelectric Nanoparticles: Dopaminergic Alterations and Their Role in Neurodegeneration and Drug Addiction. Molecules 2024; 29:3580. [PMID: 39124985 PMCID: PMC11314096 DOI: 10.3390/molecules29153580] [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: 06/30/2024] [Revised: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
Recent advancements in brain stimulation and nanomedicine have ushered in a new era of therapeutic interventions for psychiatric and neurodegenerative disorders. This review explores the cutting-edge innovations in brain stimulation techniques, including their applications in alleviating symptoms of main neurodegenerative disorders and addiction. Deep Brain Stimulation (DBS) is an FDA-approved treatment for specific neurodegenerative disorders, including Parkinson's Disease (PD), and is currently under evaluation for other conditions, such as Alzheimer's Disease. This technique has facilitated significant advancements in understanding brain electrical circuitry by enabling targeted brain stimulation and providing insights into neural network function and dysfunction. In reviewing DBS studies, this review places particular emphasis on the underlying main neurotransmitter modifications and their specific brain area location, particularly focusing on the dopaminergic system, which plays a critical role in these conditions. Furthermore, this review delves into the groundbreaking developments in nanomedicine, highlighting how nanotechnology can be utilized to target aberrant signaling in neurodegenerative diseases, with a specific focus on the dopaminergic system. The discussion extends to emerging technologies such as magnetoelectric nanoparticles (MENPs), which represent a novel intersection between nanoformulation and brain stimulation approaches. These innovative technologies offer promising avenues for enhancing the precision and effectiveness of treatments by enabling the non-invasive, targeted delivery of therapeutic agents as well as on-site, on-demand stimulation. By integrating insights from recent research and technological advances, this review aims to provide a comprehensive understanding of how brain stimulation and nanomedicine can be synergistically applied to address complex neuropsychiatric and neurodegenerative disorders, paving the way for future therapeutic strategies.
Collapse
Affiliation(s)
- Silvia Giménez
- Department of Psychobiology, Universidad de Valencia, 46010 Valencia, Spain; (S.G.); (N.A.)
| | - Alexandra Millan
- Department of Neurobiology and Neurophysiology, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain;
| | - Alba Mora-Morell
- Faculty of Biological Sciences, Universidad de Valencia, 46100 Valencia, Spain;
| | - Noa Ayuso
- Department of Psychobiology, Universidad de Valencia, 46010 Valencia, Spain; (S.G.); (N.A.)
| | - Isis Gastaldo-Jordán
- Psychiatry Service, Doctor Peset University Hospital, FISABIO, 46017 Valencia, Spain;
| | - Marta Pardo
- Department of Psychobiology, Universidad de Valencia, 46010 Valencia, Spain; (S.G.); (N.A.)
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM), 46022 Valencia, Spain
| |
Collapse
|
14
|
Akyuz E, Arulsamy A, Aslan FS, Sarisözen B, Guney B, Hekimoglu A, Yilmaz BN, Retinasamy T, Shaikh MF. An Expanded Narrative Review of Neurotransmitters on Alzheimer's Disease: The Role of Therapeutic Interventions on Neurotransmission. Mol Neurobiol 2024:10.1007/s12035-024-04333-y. [PMID: 39012443 DOI: 10.1007/s12035-024-04333-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 06/24/2024] [Indexed: 07/17/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease. The accumulation of amyloid-β (Aβ) plaques and tau neurofibrillary tangles are the key players responsible for the pathogenesis of the disease. The accumulation of Aβ plaques and tau affect the balance in chemical neurotransmitters in the brain. Thus, the current review examined the role of neurotransmitters in the pathogenesis of Alzheimer's disease and discusses the alterations in the neurochemical activity and cross talk with their receptors and transporters. In the presence of Aβ plaques and neurofibrillary tangles, changes may occur in the expression of neuronal receptors which in turn triggers excessive release of glutamate into the synaptic cleft contributing to cell death and neuronal damage. The GABAergic system may also be affected by AD pathology in a similar way. In addition, decreased receptors in the cholinergic system and dysfunction in the dopamine neurotransmission of AD pathology may also contribute to the damage to cognitive function. Moreover, the presence of deficiencies in noradrenergic neurons within the locus coeruleus in AD suggests that noradrenergic stimulation could be useful in addressing its pathophysiology. The regulation of melatonin, known for its effectiveness in enhancing cognitive function and preventing Aβ accumulation, along with the involvement of the serotonergic system and histaminergic system in cognition and memory, becomes remarkable for promoting neurotransmission in AD. Additionally, nitric oxide and adenosine-based therapeutic approaches play a protective role in AD by preventing neuroinflammation. Overall, neurotransmitter-based therapeutic strategies emerge as pivotal for addressing neurotransmitter homeostasis and neurotransmission in the context of AD. This review discussed the potential for neurotransmitter-based drugs to be effective in slowing and correcting the neurodegenerative processes in AD by targeting the neurochemical imbalance in the brain. Therefore, neurotransmitter-based drugs could serve as a future therapeutic strategy to tackle AD.
Collapse
Affiliation(s)
- Enes Akyuz
- Department of Biophysics, International School of Medicine, University of Health Sciences, Istanbul, Turkey
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
| | | | - Bugra Sarisözen
- School of Medicine, Tekirdağ Namık Kemal University, Tekirdağ, Turkey
| | - Beyzanur Guney
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | | | - Beyza Nur Yilmaz
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Thaarvena Retinasamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
- School of Dentistry and Medical Sciences, Charles Sturt University, Orange, New South Wales, 2800, Australia.
| |
Collapse
|
15
|
Gong W, Fu Y, Wu BS, Du J, Yang L, Zhang YR, Chen SD, Kang J, Mao Y, Dong Q, Tan L, Feng J, Cheng W, Yu JT. Whole-exome sequencing identifies protein-coding variants associated with brain iron in 29,828 individuals. Nat Commun 2024; 15:5540. [PMID: 38956042 PMCID: PMC11219919 DOI: 10.1038/s41467-024-49702-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/16/2024] [Indexed: 07/04/2024] Open
Abstract
Iron plays a fundamental role in multiple brain disorders. However, the genetic underpinnings of brain iron and its implications for these disorders are still lacking. Here, we conduct an exome-wide association analysis of brain iron, measured by quantitative susceptibility mapping technique, across 26 brain regions among 26,789 UK Biobank participants. We find 36 genes linked to brain iron, with 29 not being previously reported, and 16 of them can be replicated in an independent dataset with 3,039 subjects. Many of these genes are involved in iron transport and homeostasis, such as FTH1 and MLX. Several genes, while not previously connected to brain iron, are associated with iron-related brain disorders like Parkinson's (STAB1, KCNA10), Alzheimer's (SHANK1), and depression (GFAP). Mendelian randomization analysis reveals six causal relationships from regional brain iron to brain disorders, such as from the hippocampus to depression and from the substantia nigra to Parkinson's. These insights advance our understanding of the genetic architecture of brain iron and offer potential therapeutic targets for brain disorders.
Collapse
Affiliation(s)
- Weikang Gong
- School of Data Science, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Centre for Functional MRI of the Brain (FMRIB), Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, OX3 9DU, UK.
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, 266071, Qingdao, China
| | - Bang-Sheng Wu
- School of Data Science, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jingnan Du
- Department of Psychology, Center for Brain Science, Harvard University, Cambridge, MA, 02138, USA
| | - Liu Yang
- School of Data Science, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Ya-Ru Zhang
- School of Data Science, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- School of Data Science, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - JuJiao Kang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 200433, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Fudan University, 200433, Shanghai, China
| | - Ying Mao
- School of Data Science, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Qiang Dong
- School of Data Science, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, 266071, Qingdao, China
| | - Jianfeng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 200433, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Fudan University, 200433, Shanghai, China
- Department of Computer Science, University of Warwick, Coventry, UK
| | - Wei Cheng
- School of Data Science, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 200433, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Fudan University, 200433, Shanghai, China.
| | - Jin-Tai Yu
- School of Data Science, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
16
|
Hernández-Rodríguez JF, Trachioti MG, Hrbac J, Rojas D, Escarpa A, Prodromidis MI. Spark-Discharge-Activated 3D-Printed Electrochemical Sensors. Anal Chem 2024; 96:10127-10133. [PMID: 38867513 PMCID: PMC11209655 DOI: 10.1021/acs.analchem.4c01249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/11/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024]
Abstract
3D printing technology is a tremendously powerful technology to fabricate electrochemical sensing devices. However, current conductive filaments are not aimed at electrochemical applications and therefore require intense activation protocols to unleash a suitable electrochemical performance. Current activation methods based on (electro)chemical activation (using strong alkaline solutions and organic solvents and/or electrochemical treatments) or combined approaches are time-consuming and require hazardous chemicals and dedicated operator intervention. Here, pioneering spark-discharge-activated 3D-printed electrodes were developed and characterized, and it was demonstrated that their electrochemical performance was greatly improved by the effective removal of the thermoplastic support polylactic acid (PLA) as well as the formation of sponge-like and low-dimensional carbon nanostructures. This reagent-free approach consists of a direct, fast, and automatized spark discharge between the 3D-electrode and the respective graphite pencil electrode tip using a high-voltage power supply. Activated electrodes were challenged toward the simultaneous voltammetric determination of dopamine (DP) and serotonin (5-HT) in cell culture media. Spark discharge has been demonstrated as a promising approach for conductive filament activation as it is a fast, green (0.94 GREEnness Metric Approach), and automatized procedure that can be integrated into the 3D printing pipeline.
Collapse
Affiliation(s)
- Juan F. Hernández-Rodríguez
- Department
of Analytical Chemistry, Physical Chemistry and Chemical Engineering, University of Alcalá, Alcalá de Henares 28802, Madrid, Spain
| | - Maria G. Trachioti
- Department
of Chemistry, University of Ioannina, 45 110 Ioannina, Greece
| | - Jan Hrbac
- Department
of Chemistry, Masaryk University, 625 00 Brno, Czech Republic
| | - Daniel Rojas
- Department
of Analytical Chemistry, Physical Chemistry and Chemical Engineering, University of Alcalá, Alcalá de Henares 28802, Madrid, Spain
| | - Alberto Escarpa
- Department
of Analytical Chemistry, Physical Chemistry and Chemical Engineering, University of Alcalá, Alcalá de Henares 28802, Madrid, Spain
- Chemical
Research Institute “Andres M. Del Rio”, University of Alcalá, Alcalá
de Henares 28802, Madrid, Spain
| | | |
Collapse
|
17
|
List EO, Basu R, Berryman DE, Duran-Ortiz S, Martos-Moreno GÁ, Kopchick JJ. Common and uncommon mouse models of growth hormone deficiency. Endocr Rev 2024:bnae017. [PMID: 38853618 DOI: 10.1210/endrev/bnae017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/22/2024] [Accepted: 05/31/2024] [Indexed: 06/11/2024]
Abstract
Mouse models of growth hormone deficiency (GHD) have provided important tools for uncovering the various actions of GH. Nearly 100 years of research using these mouse lines has greatly enhanced our knowledge of the GH/IGF-1 axis. Some of the shared phenotypes of the five "common" mouse models of GHD include reduced body size, delayed sexual maturation, decreased fertility, reduced muscle mass, increased adiposity, and enhanced insulin sensitivity. Since these common mouse lines outlive their normal-sized littermates - and have protection from age-associated disease - they have become important fixtures in the aging field. On the other hand, the twelve "uncommon" mouse models of GHD described herein have tremendously divergent health outcomes ranging from beneficial aging phenotypes (similar to those described for the common models) to extremely detrimental features (such as improper development of the CNS, numerous sensory organ defects, and embryonic lethality). Moreover, advancements in next generation sequencing technologies have led to the identification of an expanding array of genes that are recognized as causative agents to numerous rare syndromes with concomitant GHD. Accordingly, this review provides researchers with a comprehensive up-to-date collection of the common and uncommon mouse models of GHD that have been used to study various aspects of physiology and metabolism associated with multiple forms of GHD. For each mouse line presented, the closest comparable human syndromes are discussed providing important parallels to the clinic.
Collapse
Affiliation(s)
- Edward O List
- Edison Biotechnology Institute, Ohio University, Athens Ohio, 45701, United States
- Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Athens Ohio
| | - Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens Ohio, 45701, United States
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens Ohio, 45701, United States
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens Ohio
| | - Silvana Duran-Ortiz
- Edison Biotechnology Institute, Ohio University, Athens Ohio, 45701, United States
| | - Gabriel Á Martos-Moreno
- Department of Endocrinology & Pediatrics, Hospital Infantil Universitario Niño Jesús, IIS La Princesa & Universidad Autónoma de Madrid. CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens Ohio, 45701, United States
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens Ohio
| |
Collapse
|
18
|
Bánáti D, Hellman-Regen J, Mack I, Young HA, Benton D, Eggersdorfer M, Rohn S, Dulińska-Litewka J, Krężel W, Rühl R. Defining a vitamin A5/X specific deficiency - vitamin A5/X as a critical dietary factor for mental health. INT J VITAM NUTR RES 2024; 94:443-475. [PMID: 38904956 DOI: 10.1024/0300-9831/a000808] [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: 06/22/2024]
Abstract
A healthy and balanced diet is an important factor to assure a good functioning of the central and peripheral nervous system. Retinoid X receptor (RXR)-mediated signaling was identified as an important mechanism of transmitting major diet-dependent physiological and nutritional signaling such as the control of myelination and dopamine signalling. Recently, vitamin A5/X, mainly present in vegetables as provitamin A5/X, was identified as a new concept of a vitamin which functions as the nutritional precursor for enabling RXR-mediated signaling. The active form of vitamin A5/X, 9-cis-13,14-dehydroretinoic acid (9CDHRA), induces RXR-activation, thereby acting as the central switch for enabling various heterodimer-RXR-signaling cascades involving various partner heterodimers like the fatty acid and eicosanoid receptors/peroxisome proliferator-activated receptors (PPARs), the cholesterol receptors/liver X receptors (LXRs), the vitamin D receptor (VDR), and the vitamin A(1) receptors/retinoic acid receptors (RARs). Thus, nutritional supply of vitamin A5/X might be a general nutritional-dependent switch for enabling this large cascade of hormonal signaling pathways and thus appears important to guarantee an overall organism homeostasis. RXR-mediated signaling was shown to be dependent on vitamin A5/X with direct effects for beneficial physiological and neuro-protective functions mediated systemically or directly in the brain. In summary, through control of dopamine signaling, amyloid β-clearance, neuro-protection and neuro-inflammation, the vitamin A5/X - RXR - RAR - vitamin A(1)-signaling might be "one of" or even "the" critical factor(s) necessary for good mental health, healthy brain aging, as well as for preventing drug addiction and prevention of a large array of nervous system diseases. Likewise, vitamin A5/X - RXR - non-RAR-dependent signaling relevant for myelination/re-myelination and phagocytosis/brain cleanup will contribute to such regulations too. In this review we discuss the basic scientific background, logical connections and nutritional/pharmacological expert recommendations for the nervous system especially considering the ageing brain.
Collapse
Affiliation(s)
- Diána Bánáti
- Department of Food Engineering, Faculty of Engineering, University of Szeged, Hungary
| | - Julian Hellman-Regen
- Department of Psychiatry, Charité-Campus Benjamin Franklin, Section Neurobiology, University Medicine Berlin, Germany
| | - Isabelle Mack
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tübingen, Germany
| | - Hayley A Young
- Faculty of Medicine, Health and Life Sciences, Swansea University, UK
| | - David Benton
- Faculty of Medicine, Health and Life Sciences, Swansea University, UK
| | - Manfred Eggersdorfer
- Department of Healthy Ageing, University Medical Center Groningen (UMCG), The Netherlands
| | - Sascha Rohn
- Department of Food Chemistry and Analysis, Institute of Food Technology and Food Chemistry, Technische Universität Berlin, Germany
| | | | - Wojciech Krężel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U1258, CNRS UMR 7104, Université de Strasbourg, Illkirch, France
| | | |
Collapse
|
19
|
Appiah D, Ingabire-Gasana E, Appiah L, Yang J. The Relation of Serum Vitamin C Concentrations with Alzheimer's Disease Mortality in a National Cohort of Community-Dwelling Elderly Adults. Nutrients 2024; 16:1672. [PMID: 38892605 PMCID: PMC11174700 DOI: 10.3390/nu16111672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
The relation of vitamin C with Alzheimer's disease (AD) is equivocal. The aim of this study was to assess the relation of serum vitamin C levels with AD-related mortality, and to evaluate the threshold beyond which the potential benefits of higher serum concentrations of vitamin C for AD mortality ceases. The cohort consisted of 4504 adults aged ≥60 years enrolled in the National Health and Nutrition Examination Survey who had serum measures of vitamin C and no cognitive impairment at baseline (1988-1994) and were followed-up for mortality until 2019. Vitamin C was assayed from fasting blood samples using isocratic high-performance liquid chromatography. At baseline, the mean age of participants was 70 years, with 42.7% being men. At the end of follow-up (median: 15 years), the AD mortality rate was 2.4 per 1000 person-years. In the Cox regression models, compared to participants in the lowest tertile of serum vitamin C (<0.56 mg/dL), those in the highest tertile (>0.98 mg/dL) had a lower risk of AD mortality (hazard ratio: 0.44, 95% confidence intervals: 0.25-0.77) after adjusting for sociodemographic factors, behavior/lifestyle factors, prevalent health conditions, and dietary vitamin C intake. In dose-response analysis using restricted cubic splines, vitamin C concentrations beyond 2.3 mg/dL were associated with the elevated risk of AD-related mortality. The findings from this national sample of community-dwelling elderly adults suggest that higher levels of serum vitamin C are associated with slower AD disease progression, although levels beyond the normal reference values were associated with a higher risk of AD mortality.
Collapse
Affiliation(s)
- Duke Appiah
- Department of Public Health, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | - Linda Appiah
- College of Education, Texas Tech University, Lubbock, TX 79409, USA
| | - Jeanne Yang
- School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
20
|
Pan X, Tan X, McDonald J, Kaminga AC, Chen Y, Dai F, Qiu J, Zhao K, Peng Y. Chemokines in diabetic eye disease. Diabetol Metab Syndr 2024; 16:115. [PMID: 38790059 PMCID: PMC11127334 DOI: 10.1186/s13098-024-01297-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 02/20/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Diabetic eye disease is a common micro-vascular complication of diabetes and a leading cause of decreased vision and blindness in people of working age worldwide.Although previous studies have shown that chemokines system may be a player in pathogenesis of diabetic eye disease, it is unclear which chemokines play the most important role.To date, there is no meta-analysis which has investigated the role of chemokines in diabetic eye disease.We hope this study will contribute to a better understanding of both the signaling pathways of the chemokines in the pathophysiological process, and more reliable therapeutic targets for diabetic eye disease. METHODS Embase, PubMed, Web of Science and Cochrane Library systematically searched for relevant studies from inception to Sep 1, 2023. A random-effect model was used and standardized mean differences (SMDs) and 95% confidence intervals (CIs) were calculated to summarize the associated measure between chemokines concentrations and diabetic eye disease. Network meta-analysis to rank chemokines-effect values according to ranked probabilities. RESULTS A total of 33 different chemokines involving 11,465 subjects (6559 cases and 4906 controls) were included in the meta-analysis. Results of the meta-analysis showed that concentrations of CC and CXC chemokines in the diabetic eye disease patients were significantly higher than those in the controls. Moreover, network meta-analysis showed that the effect of CCL8, CCL2, CXCL8 and CXCL10 were ranked highest in terms of probabilities. Concentrations of CCL8, CCL2, CXCL8 and CXCL10 may be associated with diabetic eye disease, especially in diabetic retinopathy and diabetic macular edema. CONCLUSION Our study suggests that CCL2 and CXCL8 may play key roles in pathogenesis of diabetic eye disease. Future research should explore putative mechanisms underlying these links, with the commitment to develop novel prophylactic and therapeutic for diabetic eye disease.
Collapse
Affiliation(s)
- Xiongfeng Pan
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, 86 Ziyuan Rd, Changsha, Hunan, People's Republic of China, 410007.
- The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University, Changsha, China.
| | - Xinrui Tan
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Judy McDonald
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, ON, Canada
| | | | - Yuyao Chen
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Feizhao Dai
- Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jun Qiu
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, 86 Ziyuan Rd, Changsha, Hunan, People's Republic of China, 410007
| | - Kunyan Zhao
- School of Public Health, University of South China, Hengyang, China
| | - Yunlong Peng
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
21
|
Rajarathinam T, Jayaraman S, Seol J, Lee J, Chang SC. Utilizing a Disposable Sensor with Polyaniline-Doped Multi-Walled Carbon Nanotubes to Enable Dopamine Detection in Ex Vivo Mouse Brain Tissue Homogenates. BIOSENSORS 2024; 14:262. [PMID: 38920566 PMCID: PMC11201478 DOI: 10.3390/bios14060262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 06/27/2024]
Abstract
Disposable sensors are inexpensive, user-friendly sensing tools designed for rapid single-point measurements of a target. Disposable sensors have become more and more essential as diagnostic tools due to the growing demand for quick, easy-to-access, and reliable information related to the target. Dopamine (DA), a prevalent catecholamine neurotransmitter in the human brain, is associated with central nervous system activities and directly promotes neuronal communication. For the sensitive and selective estimation of DA, an enzyme-free amperometric sensor based on polyaniline-doped multi-walled carbon nanotubes (PANI-MWCNTs) drop-coated disposable screen-printed carbon electrodes (SPCEs) was fabricated. This PANI-MWCNTs-2/SPCE sensor boasts exceptional accuracy and sensitivity when working directly with ex vivo mouse brain homogenates. The sensor exhibited a detection limit of 0.05 μM (S/N = 3), and a wide linear range from 1.0 to 200 μM. The sensor's high selectivity to DA amidst other endogenous interferents was recognized. Since the constructed sensor is enzyme-free yet biocompatible, it exhibited high stability in DA detection using ex vivo mouse brain homogenates extracted from both Parkinson's disease and control mice models. This research thus presents new insights into understanding DA release dynamics at the tissue level in both of these models.
Collapse
Affiliation(s)
- Thenmozhi Rajarathinam
- Engineering Research Center for Color-Modulated Extra-Sensory Perception Technology, Pusan National University, Busan 46241, Republic of Korea;
| | - Sivaguru Jayaraman
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea;
| | - Jaeheon Seol
- BIT Convergence-Based Innovative Drug Development Targeting Metainflammation, Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea; (J.S.); (J.L.)
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Jaewon Lee
- BIT Convergence-Based Innovative Drug Development Targeting Metainflammation, Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea; (J.S.); (J.L.)
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Seung-Cheol Chang
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea;
| |
Collapse
|
22
|
Kim J, Yoo H, Woo S, Oh SS. Aptasensor-encapsulating semi-permeable proteinosomes for direct target detection in non-treated biofluids. Biosens Bioelectron 2024; 251:116062. [PMID: 38350238 DOI: 10.1016/j.bios.2024.116062] [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: 10/30/2023] [Revised: 01/12/2024] [Accepted: 01/22/2024] [Indexed: 02/15/2024]
Abstract
Detecting biomarkers in biofluids directly without sample treatments makes molecular diagnostics faster and more efficient. Aptasensors, the nucleic acid-based molecular biosensors, can detect a wide range of target molecules, but their susceptibility to degradation and aggregation by nucleases and charged proteins, respectively, limits their direct use in clinical samples. In this work, we demonstrate that when aptasensors are encapsulated in proteinosomes, the protein-based liposome mimics, clinically important small molecules can be sensitively and selectively detected in non-treated specimens, such as 100 % unpurified serum. As serum albumin is used to form the membrane, the nanomeshed proteinosomes become semi-permeable and antifouling, which enables exclusive admission of small molecules while blocking unwanted large proteins. Consequently, the enclosed aptasensors can maintain close-to-optimal performance for target binding, and nucleolytic degradation and electrostatic aggregation are effectively suppressed. Three different structure-switching aptamers specific for estradiol, dopamine, and cocaine, respectively, are demonstrated to fully conserve their high affinities and specificities inside the microcapsules. The shielding effect of proteinosomes is indeed exceptional; the enclosed DNA aptasensors remain completely intact over 18 h in serum and even in an extremely concentrated DNase solution (1 mg/ml, ∼300,000× the serum level). Moreover, the proteinosome-mediated compartmentalization enables independent operation of multiple aptasensors in the same mixture. Hence, simultaneous real-time sensing of two different targets is demonstrated with different operation modes, 'recording' target appearance and 'reporting' target concentration changes. This work is the first demonstration of small-molecule-specific aptasensors operating with optimal performance in serum environments and will find promising applications in molecular diagnostics.
Collapse
Affiliation(s)
- Jinmin Kim
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, South Korea
| | - Hyebin Yoo
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, South Korea
| | - Sungwook Woo
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, South Korea.
| | - Seung Soo Oh
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, South Korea; Institute for Convergence Research and Education in Advanced Technology (I-CREATE), Yonsei University, Incheon, 21983, South Korea.
| |
Collapse
|
23
|
Fakih N, Fakhoury M. Alzheimer Disease-Link With Major Depressive Disorder and Efficacy of Antidepressants in Modifying its Trajectory. J Psychiatr Pract 2024; 30:181-191. [PMID: 38819242 DOI: 10.1097/pra.0000000000000779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Alzheimer disease (AD) is a devastating neurodegenerative disorder that affects millions of individuals worldwide, with no effective cure. The main symptoms include learning and memory loss, and the inability to carry out the simplest tasks, significantly affecting patients' quality of life. Over the past few years, tremendous progress has been made in research demonstrating a link between AD and major depressive disorder (MDD). Evidence suggests that MDD is commonly associated with AD and that it can serve as a precipitating factor for this disease. Antidepressants such as selective serotonin reuptake inhibitors, which are the first line of treatment for MDD, have shown great promise in the treatment of depression in AD, although their effectiveness remains controversial. The goal of this review is to summarize current knowledge regarding the association between AD, MDD, and antidepressant treatment. It first provides an overview of the interaction between AD and MDD at the level of genes, brain regions, neurotransmitter systems, and neuroinflammatory markers. The review then presents current evidence regarding the effectiveness of various antidepressants for AD-related pathophysiology and then finally discusses current limitations, challenges, and future directions.
Collapse
Affiliation(s)
- Nour Fakih
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | | |
Collapse
|
24
|
Liu C, Huang D, Sheng X, Zhu J, Dong S, Chen S, Wang Y, Tang A, Duan R, Yang Z, Bai J, Zheng Y. Integrated physiological, intestinal microbiota, and metabolomic responses of adult zebrafish (Danio rerio) to subacute exposure to antimony at environmentally relevant concentrations. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 277:116326. [PMID: 38640800 DOI: 10.1016/j.ecoenv.2024.116326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/30/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024]
Abstract
The available information regarding the impact of antimony (Sb), a novel environmental pollutant, on the intestinal microbiota and host health is limited. In this study, we conducted physiological characterizations to investigate the response of adult zebrafish to different environmental concentrations (0, 30, 300, and 3000 µg/L) of Sb over a period of 14 days. Biochemical and pathological changes demonstrated that Sb effectively compromised the integrity of the intestinal physical barrier and induced inflammatory responses as well as oxidative stress. Analysis of both intestinal microbial community and metabolome revealed that exposure to 0 and 30 µg/L of Sb resulted in similar microbiota structures; however, exposure to 300 µg/L altered microbial communities' composition (e.g., a decline in genus Cetobacterium and an increase in Vibrio). Furthermore, exposure to 300 µg/L significantly decreased levels of bile acids and glycerophospholipids while triggering intestinal inflammation but activating self-protective mechanisms such as antibiotic presence. Notably, even exposure to 30 µg/L of Sb can trigger dysbiosis of intestinal microbiota and metabolites, potentially impacting fish health through the "microbiota-intestine-brain axis" and contributing to disease initiation. This study provides valuable insights into toxicity-related information concerning environmental impacts of Sb on aquatic organisms with significant implications for developing management strategies.
Collapse
Affiliation(s)
- Can Liu
- Hunan University of Humanities, Science and Technology, Loudi, Hunan 417000, China; Hunan Provincial Collaborative Innovation Center for Field Weeds Control, Hunan University of Humanities, Science and Technology, Loudi, Hunan 417000, China
| | - Dongmei Huang
- Hunan University of Humanities, Science and Technology, Loudi, Hunan 417000, China
| | - Xiangquan Sheng
- Hunan University of Humanities, Science and Technology, Loudi, Hunan 417000, China
| | - Jianzhong Zhu
- Hunan University of Humanities, Science and Technology, Loudi, Hunan 417000, China
| | - Si Dong
- Hunan University of Humanities, Science and Technology, Loudi, Hunan 417000, China
| | - Song Chen
- Hunan University of Humanities, Science and Technology, Loudi, Hunan 417000, China
| | - Yaying Wang
- Hunan University of Humanities, Science and Technology, Loudi, Hunan 417000, China
| | - Ao Tang
- Hunan University of Humanities, Science and Technology, Loudi, Hunan 417000, China
| | - Renyan Duan
- Hunan University of Humanities, Science and Technology, Loudi, Hunan 417000, China
| | - Zeliang Yang
- Hunan University of Humanities, Science and Technology, Loudi, Hunan 417000, China
| | - Jing Bai
- Hunan University of Humanities, Science and Technology, Loudi, Hunan 417000, China.
| | - Yu Zheng
- Hunan University of Humanities, Science and Technology, Loudi, Hunan 417000, China; Hunan Provincial Collaborative Innovation Center for Field Weeds Control, Hunan University of Humanities, Science and Technology, Loudi, Hunan 417000, China.
| |
Collapse
|
25
|
Onisiforou A, Christodoulou CC, Zamba-Papanicolaou E, Zanos P, Georgiou P. Transcriptomic analysis reveals sex-specific patterns in the hippocampus in Alzheimer's disease. Front Endocrinol (Lausanne) 2024; 15:1345498. [PMID: 38689734 PMCID: PMC11058985 DOI: 10.3389/fendo.2024.1345498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/02/2024] [Indexed: 05/02/2024] Open
Abstract
Background The hippocampus, vital for memory and learning, is among the first brain regions affected in Alzheimer's Disease (AD) and exhibits adult neurogenesis. Women face twice the risk of developing AD compare to men, making it crucial to understand sex differences in hippocampal function for comprehending AD susceptibility. Methods We conducted a comprehensive analysis of bulk mRNA postmortem samples from the whole hippocampus (GSE48350, GSE5281) and its CA1 and CA3 subfields (GSE29378). Our aim was to perform a comparative molecular signatures analysis, investigating sex-specific differences and similarities in the hippocampus and its subfields in AD. This involved comparing the gene expression profiles among: (a) male controls (M-controls) vs. female controls (F-controls), (b) females with AD (F-AD) vs. F-controls, (c) males with AD (M-AD) vs. M-controls, and (d) M-AD vs. F-AD. Furthermore, we identified AD susceptibility genes interacting with key targets of menopause hormone replacement drugs, specifically the ESR1 and ESR2 genes, along with GPER1. Results The hippocampal analysis revealed contrasting patterns between M-AD vs. M-controls and F-AD vs. F-controls, as well as M-controls vs. F-controls. Notably, BACE1, a key enzyme linked to amyloid-beta production in AD pathology, was found to be upregulated in M-controls compared to F-controls in both CA1 and CA3 hippocampal subfields. In M-AD vs. M-controls, the GABAergic synapse was downregulated, and the Estrogen signaling pathway was upregulated in both subfields, unlike in F-AD vs. F-controls. Analysis of the whole hippocampus also revealed upregulation of the GABAergic synapse in F-AD vs. F-controls. While direct comparison of M-AD vs. F-AD, revealed a small upregulation of the ESR1 gene in the CA1 subfield of males. Conversely, F-AD vs. F-controls exhibited downregulation of the Dopaminergic synapse in both subfields, while the Calcium signaling pathway showed mixed regulation, being upregulated in CA1 but downregulated in CA3, unlike in M-AD vs. M-controls. The upregulated Estrogen signaling pathway in M-AD, suggests a compensatory response to neurodegenerative specifically in males with AD. Our results also identified potential susceptibility genes interacting with ESR1 and ESR2, including MAPK1, IGF1, AKT1, TP53 and CD44. Conclusion These findings underscore the importance of sex-specific disease mechanisms in AD pathogenesis. Region-specific analysis offers a more detailed examination of localized changes in the hippocampus, enabling to capture sex-specific molecular patterns in AD susceptibility and progression.
Collapse
Affiliation(s)
- Anna Onisiforou
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, Nicosia, Cyprus
| | | | | | - Panos Zanos
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, Nicosia, Cyprus
| | - Polymnia Georgiou
- Laboratory of Epigenetics and Gene Regulation, Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
- Psychoneuroendocrinology Laboratory, Department of Psychology, University of Wisconsin Milwaukee, Milwaukee, WI, United States
| |
Collapse
|
26
|
Ceylan B, Düz E, Çakir T. Personalized Protein-Protein Interaction Networks Towards Unraveling the Molecular Mechanisms of Alzheimer's Disease. Mol Neurobiol 2024; 61:2120-2135. [PMID: 37855983 DOI: 10.1007/s12035-023-03690-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/02/2023] [Indexed: 10/20/2023]
Abstract
Alzheimer's disease (AD) is a highly heterogenous neurodegenerative disease, and several omic-based datasets were generated in the last decade from the patients with the disease. However, the vast majority of studies evaluate these datasets in bulk by considering all the patients as a single group, which obscures the molecular differences resulting from the heterogeneous nature of the disease. In this study, we adopted a personalized approach and analyzed the transcriptome data from 403 patients individually by mapping the data on a human protein-protein interaction network. Patient-specific subnetworks were discovered and analyzed in terms of the genes in the subnetworks, enriched functional terms, and known AD genes. We identified several affected pathways that could not be captured by the bulk comparison. We also showed that our personalized findings point to patterns of alterations consistent with the recently suggested AD subtypes.
Collapse
Affiliation(s)
- Betül Ceylan
- Department of Bioengineering, Gebze Technical University, 41400, Gebze, Kocaeli, Turkey
| | - Elif Düz
- Department of Bioengineering, Gebze Technical University, 41400, Gebze, Kocaeli, Turkey
| | - Tunahan Çakir
- Department of Bioengineering, Gebze Technical University, 41400, Gebze, Kocaeli, Turkey.
| |
Collapse
|
27
|
Elmers J, Colzato LS, Ziemssen F, Ziemssen T, Beste C. Optical coherence tomography as a potential surrogate marker of dopaminergic modulation across the life span. Ageing Res Rev 2024; 96:102280. [PMID: 38518921 DOI: 10.1016/j.arr.2024.102280] [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: 09/27/2023] [Revised: 02/02/2024] [Accepted: 03/18/2024] [Indexed: 03/24/2024]
Abstract
The retina has been considered a "window to the brain" and shares similar innervation by the dopaminergic system with the cortex in terms of an unequal distribution of D1 and D2 receptors. Here, we provide a comprehensive overview that Optical Coherence Tomography (OCT), a non-invasive imaging technique, which provides an "in vivo" representation of the retina, shows promise to be used as a surrogate marker of dopaminergic neuromodulation in cognition. Overall, most evidence supports reduced retinal thickness in individuals with dopaminergic dysregulation (e.g., patients with Parkinson's Disease, non-demented older adults) and with poor cognitive functioning. By using the theoretical framework of metacontrol, we derive hypotheses that retinal thinning associated to decreased dopamine (DA) levels affecting D1 families, might lead to a decrease in the signal-to-noise ratio (SNR) affecting cognitive persistence (depending on D1-modulated DA activity) but not cognitive flexibility (depending on D2-modulated DA activity). We argue that the use of OCT parameters might not only be an insightful for cognitive neuroscience research, but also a potentially effective tool for individualized medicine with a focus on cognition. As our society progressively ages in the forthcoming years and decades, the preservation of cognitive abilities and promoting healthy aging will hold of crucial significance. OCT has the potential to function as a swift, non-invasive, and economical method for promptly recognizing individuals with a heightened vulnerability to cognitive deterioration throughout all stages of life.
Collapse
Affiliation(s)
- Julia Elmers
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany; Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Lorenza S Colzato
- Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan, China
| | - Focke Ziemssen
- Ophthalmological Clinic, University Clinic Leipzig, Germany
| | - Tjalf Ziemssen
- Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany; Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan, China.
| |
Collapse
|
28
|
Alam F, Ashfaq Ahmed M, Jalal AH, Siddiquee I, Adury RZ, Hossain GMM, Pala N. Recent Progress and Challenges of Implantable Biodegradable Biosensors. MICROMACHINES 2024; 15:475. [PMID: 38675286 PMCID: PMC11051912 DOI: 10.3390/mi15040475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024]
Abstract
Implantable biosensors have evolved to the cutting-edge technology of personalized health care and provide promise for future directions in precision medicine. This is the reason why these devices stand to revolutionize our approach to health and disease management and offer insights into our bodily functions in ways that have never been possible before. This review article tries to delve into the important developments, new materials, and multifarious applications of these biosensors, along with a frank discussion on the challenges that the devices will face in their clinical deployment. In addition, techniques that have been employed for the improvement of the sensitivity and specificity of the biosensors alike are focused on in this article, like new biomarkers and advanced computational and data communicational models. A significant challenge of miniaturized in situ implants is that they need to be removed after serving their purpose. Surgical expulsion provokes discomfort to patients, potentially leading to post-operative complications. Therefore, the biodegradability of implants is an alternative method for removal through natural biological processes. This includes biocompatible materials to develop sensors that remain in the body over longer periods with a much-reduced immune response and better device longevity. However, the biodegradability of implantable sensors is still in its infancy compared to conventional non-biodegradable ones. Sensor design, morphology, fabrication, power, electronics, and data transmission all play a pivotal role in developing medically approved implantable biodegradable biosensors. Advanced material science and nanotechnology extended the capacity of different research groups to implement novel courses of action to design implantable and biodegradable sensor components. But the actualization of such potential for the transformative nature of the health sector, in the first place, will have to surmount the challenges related to biofouling, managing power, guaranteeing data security, and meeting today's rules and regulations. Solving these problems will, therefore, not only enhance the performance and reliability of implantable biodegradable biosensors but also facilitate the translation of laboratory development into clinics, serving patients worldwide in their better disease management and personalized therapeutic interventions.
Collapse
Affiliation(s)
- Fahmida Alam
- Department of Electrical and Computer Engineering, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA; (A.H.J.); (G.M.M.H.)
| | | | - Ahmed Hasnain Jalal
- Department of Electrical and Computer Engineering, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA; (A.H.J.); (G.M.M.H.)
| | - Ishrak Siddiquee
- Institute of Microsystems Technology, University of South-Eastern Norway, Horten, 3184 Vestfold, Norway;
| | - Rabeya Zinnat Adury
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL 32611, USA;
| | - G M Mehedi Hossain
- Department of Electrical and Computer Engineering, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA; (A.H.J.); (G.M.M.H.)
| | - Nezih Pala
- Department of Electrical and Computer Engineering, Florida International University, Miami, FL 33174, USA;
| |
Collapse
|
29
|
Li W, Chang C, Kundu S, Long Q. Accounting for network noise in graph-guided Bayesian modeling of structured high-dimensional data. Biometrics 2024; 80:ujae012. [PMID: 38483282 PMCID: PMC10938547 DOI: 10.1093/biomtc/ujae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/31/2023] [Accepted: 02/14/2024] [Indexed: 03/17/2024]
Abstract
There is a growing body of literature on knowledge-guided statistical learning methods for analysis of structured high-dimensional data (such as genomic and transcriptomic data) that can incorporate knowledge of underlying networks derived from functional genomics and functional proteomics. These methods have been shown to improve variable selection and prediction accuracy and yield more interpretable results. However, these methods typically use graphs extracted from existing databases or rely on subject matter expertise, which are known to be incomplete and may contain false edges. To address this gap, we propose a graph-guided Bayesian modeling framework to account for network noise in regression models involving structured high-dimensional predictors. Specifically, we use 2 sources of network information, including the noisy graph extracted from existing databases and the estimated graph from observed predictors in the dataset at hand, to inform the model for the true underlying network via a latent scale modeling framework. This model is coupled with the Bayesian regression model with structured high-dimensional predictors involving an adaptive structured shrinkage prior. We develop an efficient Markov chain Monte Carlo algorithm for posterior sampling. We demonstrate the advantages of our method over existing methods in simulations, and through analyses of a genomics dataset and another proteomics dataset for Alzheimer's disease.
Collapse
Affiliation(s)
- Wenrui Li
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, PA 19104, United States
| | - Changgee Chang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Suprateek Kundu
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Qi Long
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, PA 19104, United States
| |
Collapse
|
30
|
Esmaeili A, Dismuke-Greer C, Pogoda TK, Amuan ME, Garcia C, Del Negro A, Myers M, Kennedy E, Cifu D, Pugh MJ. Cannabis use disorder contributes to cognitive dysfunction in Veterans with traumatic brain injury. Front Neurol 2024; 15:1261249. [PMID: 38292293 PMCID: PMC10824930 DOI: 10.3389/fneur.2024.1261249] [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: 10/31/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2024] Open
Abstract
Background While emerging evidence supports a link between traumatic brain injury (TBI) and progressive cognitive dysfunction in Veterans, there is insufficient information on the impact of cannabis use disorder (CUD) on long-term cognitive disorders. This study aimed to examine the incidences of cognitive disorders in Veterans with TBI and CUD and to evaluate their relationship. Methods This retrospective cohort study used the US Department of Veterans Affairs and Department of Defense administrative data from the Long-term Impact of Military-Relevant Brain Injury Consortium-Chronic Effects of Neurotrauma Consortium Phenotype study. Diagnoses suggesting cognitive disorders after a TBI index date were identified using inpatient and outpatient data from 2003 to 2022. We compared the differential cognitive disorders incidence in Veterans who had the following: (1) no CUD or TBI (control group), (2) CUD only, (3) TBI only, and (4) comorbid CUD+TBI. Kaplan-Meier analyses were used to estimate the overall cognitive disorders incidence in the above study groups. The crude and adjusted Cox proportional hazards models were used to estimate crude and adjusted hazard ratios (HRs) for cognitive disorders. Results A total of 1,560,556 Veterans [82.32% male, median (IQR) age at the time of TBI, 34.51 (11.29) years, and 61.35% white] were evaluated. The cognitive disorder incidence rates were estimated as 0.68 (95% CI, 0.62, 0.75) for CUD only and 1.03 (95% CI, 1.00, 1.06) for TBI only per 10,000 person-months of observations, with the highest estimated cognitive disorder incidence observed in participants with both TBI and CUD [1.83 (95% CI, 1.72, 1.95)]. Relative to the control group, the highest hazard of cognitive disorders was observed in Veterans with CUD+TBI [hazard ratio (HR), 3.26; 95% CI, 2.91, 3.65], followed by those with TBI only (2.32; 95 CI%, 2.13, 2.53) and with CUD (1.79; 95 CI%, 1.60, 2.00). Of note, in the CUD only subgroup, we also observed the highest risk of an early onset cognitive disorder other than Alzheimer's disease and Frontotemporal dementia. Discussion The results of this analysis suggest that individuals with comorbid TBI and CUD may be at increased risk for early onset cognitive disorders, including dementia.
Collapse
Affiliation(s)
- Aryan Esmaeili
- Health Economics Resource Center (HERC), Ci2i, VA Palo Alto Health Care System, Menlo Park, CA, United States
| | - Clara Dismuke-Greer
- Health Economics Resource Center (HERC), Ci2i, VA Palo Alto Health Care System, Menlo Park, CA, United States
| | - Terri K. Pogoda
- Center for Healthcare Organization and Implementation Research, VA Boston Healthcare System, Boston, MA, United States
- Boston University School of Public Health, Boston, MA, United States
| | - Megan E. Amuan
- Informatics, Decision-Enhancement, and Analytic Sciences Center of Innovation, VA Salt Lake City Health Care System, Salt Lake City, UT, United States
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Carla Garcia
- Health Economics Resource Center (HERC), Ci2i, VA Palo Alto Health Care System, Menlo Park, CA, United States
| | - Ariana Del Negro
- Health Economics Resource Center (HERC), Ci2i, VA Palo Alto Health Care System, Menlo Park, CA, United States
| | - Maddy Myers
- Informatics, Decision-Enhancement, and Analytic Sciences Center of Innovation, VA Salt Lake City Health Care System, Salt Lake City, UT, United States
| | - Eamonn Kennedy
- Informatics, Decision-Enhancement, and Analytic Sciences Center of Innovation, VA Salt Lake City Health Care System, Salt Lake City, UT, United States
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - David Cifu
- Department of Physical Medicine and Rehabilitation, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Mary Jo Pugh
- Informatics, Decision-Enhancement, and Analytic Sciences Center of Innovation, VA Salt Lake City Health Care System, Salt Lake City, UT, United States
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
31
|
Kumar S, Sharma R, Singh D, Awasthi A, Kumar V, Singh K. Tungsten sulphide decorated carbon nanotube based electroanalytical sensing of neurotransmitter dopamine. Electrochim Acta 2024; 475:143584. [DOI: 10.1016/j.electacta.2023.143584] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
32
|
Cannavacciuolo A, Paparella G, Salzillo M, Colella D, Canevelli M, Costa D, Birreci D, Angelini L, Guerra A, Ricciardi L, Bruno G, Berardelli A, Bologna M. Facial emotion expressivity in patients with Parkinson's and Alzheimer's disease. J Neural Transm (Vienna) 2024; 131:31-41. [PMID: 37804428 PMCID: PMC10770202 DOI: 10.1007/s00702-023-02699-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/09/2023] [Indexed: 10/09/2023]
Abstract
Parkinson's disease (PD) and Alzheimer's disease (AD) are neurodegenerative disorders with some overlapping clinical features. Hypomimia (reduced facial expressivity) is a prominent sign of PD and it is also present in AD. However, no study has experimentally assessed hypomimia in AD and compared facial expressivity between PD and AD patients. We compared facial emotion expressivity in patients with PD, AD, and healthy controls (HCs). Twenty-four PD patients, 24 AD patients and 24 HCs were videotaped during neutral facial expressions and while posing six facial emotions (anger, surprise, disgust, fear, happiness, and sadness). Fifteen raters were asked to evaluate the videos using MDS-UPDRS-III (item 3.2) and to identify the corresponding emotion from a seven-forced-choice response format. We measured the percentage of accuracy, the reaction time (RT), and the confidence level (CL) in the perceived accuracy of the raters' responses. We found the highest MDS-UPDRS 3.2 scores in PD, and higher in AD than HCs. When evaluating the posed expression captures, raters identified a lower percentage of correct answers in the PD and AD groups than HCs. There was no difference in raters' response accuracy between the PD and AD. No difference was observed in RT and CL data between groups. Hypomimia in patients correlated positively with the global MDS-UPDRS-III and negatively with Mini Mental State Examination scores. PD and AD patients have a similar pattern of reduced facial emotion expressivity compared to controls. These findings hold potential pathophysiological and clinical implications.
Collapse
Affiliation(s)
| | - Giulia Paparella
- IRCCS Neuromed Pozzilli (IS), Pozzilli, Italy
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy
| | - Martina Salzillo
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy
| | - Donato Colella
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy
| | - Marco Canevelli
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Davide Costa
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy
| | - Daniele Birreci
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy
| | - Luca Angelini
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy
| | - Andrea Guerra
- Parkinson and Movement Disorder Unit, Study Center on Neurodegeneration (CESNE), Department of Neuroscience, University of Padua, Padua, Italy
| | - Lucia Ricciardi
- St George's, University of London and St George's University Hospitals NHS Foundation Trust, Institute of Molecular and Clinical Sciences, Neurosciences Research Centre, Cranmer Terrace, London, SW17 0QT, UK
| | - Giuseppe Bruno
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy
| | - Alfredo Berardelli
- IRCCS Neuromed Pozzilli (IS), Pozzilli, Italy
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy
| | - Matteo Bologna
- IRCCS Neuromed Pozzilli (IS), Pozzilli, Italy.
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy.
| |
Collapse
|
33
|
Paul D, Agrawal R, Singh S. Alzheimer's disease and clinical trials. J Basic Clin Physiol Pharmacol 2024; 35:31-44. [PMID: 38491747 DOI: 10.1515/jbcpp-2023-0264] [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: 12/29/2023] [Accepted: 02/28/2024] [Indexed: 03/18/2024]
Abstract
Alzheimer's disease (AD) is spreading its root disproportionately among the worldwide population. Many genes have been identified as the hallmarks of AD. Based upon the knowledge, many clinical trials have been designed and conducted. Attempts have been made to alleviate the pathology associated with AD by targeting the molecular products of these genes. Irrespective of the understanding on the genetic component of AD, many clinical trials have failed and imposed greater challenges on the path of drug discovery. Therefore, this review aims to identify research and review articles to pinpoint the limitations of drug candidates (thiethylperazine, CT1812, crenezumab, CNP520, and lecanemab), which are under or withdrawn from clinical trials. Thorough analysis of the cross-talk pathways led to the identification of many confounding factors, which could interfere with the success of clinical trials with drug candidates such as thiethylperazine, CT1812, crenezumab, and CNP520. Though these drug candidates were enrolled in clinical trials, yet literature review shows many limitations. These limitations raise many questions on the rationale behind the enrollments of these drug candidates in clinical trials. A meticulous prior assessment of the outcome of clinical studies may stop risky clinical trials at their inceptions. This may save time, money, and resources.
Collapse
Affiliation(s)
- Deepraj Paul
- Department of Pharmacology, 621320 College of Pharmacy JSS Academy of Technical Education , Noida, Uttar Pradesh, India
| | - Rohini Agrawal
- Department of Pharmacology, 621320 College of Pharmacy JSS Academy of Technical Education , Noida, Uttar Pradesh, India
| | - Swati Singh
- Department of Pharmacology, 621320 College of Pharmacy JSS Academy of Technical Education , Noida, Uttar Pradesh, India
| |
Collapse
|
34
|
Thomas J, Wilson S. Molecular and Therapeutic Targets for Amyloid-beta Plaques in Alzheimer's Disease: A Review Study. Basic Clin Neurosci 2024; 15:1-26. [PMID: 39291090 PMCID: PMC11403107 DOI: 10.32598/bcn.2021.3522.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/06/2021] [Accepted: 09/06/2021] [Indexed: 09/19/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive loss of cognition and a gradual decrease in memory. Although AD is considered the most persistent form of dementia and a global concern, no complete cure or agents that can completely halt the progression of AD have been found. In the past years, significant progress has been made in understanding the cellular and molecular changes associated with AD, and numerous drug targets have been identified for the development of drugs for this disease. Amyloid-beta (Aβ) plaques and neurofibrillary tangles (NFT) are the major attributes of AD. Symptomatic relief is the only possible treatment available at present and a disease-modifying drug is of utmost importance. The development of drugs that can inhibit different targets responsible for the formation of plaques is a potential area in AD research. This review is not a complete list of all possible targets for AD but serves to highlight the targets related to Aβ pathology and pathways concerned with the formation of Aβ fragments. This shall serve as a prospect in the identification of Aβ plaque inhibitors and pave the strategies for newer drug treatments. Nevertheless, substantial research is done in this area but to bridle, the clinical difficulty remains a concern.
Collapse
Affiliation(s)
- Jaya Thomas
- Department of Pharmacology, School of Pharmacy University of Amrita Vishwavidyapeetham, Guntur, India
| | - Samson Wilson
- University of Amrita Vishwavidyapeetham, Coimbatore, India
| |
Collapse
|
35
|
Almahasneh F, Gerges RH, Abu-El-Rub E, Khasawneh RR. Nicotine Abuse and Neurodegeneration: Novel Pharmacogenetic Targets to Aid Quitting and Reduce the Risk of Dementia. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:2-8. [PMID: 36803746 DOI: 10.2174/1871527322666230220121655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/23/2022] [Accepted: 01/11/2023] [Indexed: 02/22/2023]
Abstract
Nicotine dependence has deleterious neurological impacts. Previous studies found an association between cigarette smoking and accelerating age-related thinning of the brain's cortex and subsequent cognitive decline. Smoking is considered the third most common risk factor for dementia, which prompted the inclusion of smoking cessation in dementia prevention strategies. Traditional pharmacologic options for smoking cessation include nicotine transdermal patches, bupropion and varenicline. However, based on smokers' genetic makeup, pharmacogenetics can be used to develop novel therapies to replace these traditional approaches. Genetic variability of cytochrome P450 2A6 has a major impact on smokers' behavior and their response to quitting therapies. Gene polymorphism in nicotinic acetylcholine receptor subunits also has a great influence on the ability to quit smoking. In addition, polymorphism of certain nicotinic acetylcholine receptors was found to affect the risk of dementia and the impact of tobacco smoking on the development of Alzheimer's disease. Nicotine dependence involves the activation of pleasure response through the stimulation of dopamine release. Central dopamine receptors, catechol-o-methyltransferase and the dopamine transporter protein, regulate synaptic dopamine levels. The genes of these molecules are potential targets for novel smoking cessation drugs. Pharmacogenetic studies of smoking cessation also investigated other molecules, such as ANKK1 and dopamine-beta-hydroxylase (DBH). In this perspective article, we aim to highlight the promising role of pharmacogenetics in the development of effective drugs for smoking cessation, which can increase the success rate of smoking quitting plans and ultimately reduce the incidence of neurodegeneration and dementia.
Collapse
Affiliation(s)
- Fatimah Almahasneh
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Romany H Gerges
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ejlal Abu-El-Rub
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Ramada R Khasawneh
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| |
Collapse
|
36
|
Tumurbaatar B, Fracassi A, Scaduto P, Guptarak J, Woltjer R, Jupiter D, Taglialatela G. Preserved autophagy in cognitively intact non-demented individuals with Alzheimer's neuropathology. Alzheimers Dement 2023; 19:5355-5370. [PMID: 37191183 PMCID: PMC10651802 DOI: 10.1002/alz.13074] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/27/2023] [Accepted: 03/08/2023] [Indexed: 05/17/2023]
Abstract
INTRODUCTION Growing evidence supports that dysfunctional autophagy, the major cell mechanism responsible for removing protein aggregates and a route of clearance for Tau in healthy neurons, is a major finding in demented Alzheimer's disease (AD) patients. However, the association of autophagy with maintenance of cognitive integrity in resilient individuals who have AD neuropathology but remain non-demented (NDAN) has not been evaluated. METHODS Using post mortem brain samples from age-matched healthy control, AD, and NDAN subjects, we evaluated autophagy in relation to Tau pathology using Western blot, immunofluorescence and RNA-seq. RESULTS Compared to AD patients, NDAN subjects had preserved autophagy and reduced tauopathy. Furthermore, expression of autophagy genes and AD-related proteins were significantly associated in NDAN compared to AD and control subjects. DISCUSSION Our results suggest preserved autophagy is a protective mechanism that maintains cognitive integrity in NDAN individuals. This novel observation supports the potential of autophagy-inducing strategies in AD therapeutics. HIGHLIGHTS NDAN subjects have preserved autophagic protein levels comparable with control subjects. Compared to control subjects, NDAN subjects have significantly reduced Tau oligomers and PHF Tau phosphorylation at synapses that negatively correlate with autophagy markers. Transcription of autophagy genes strongly associates with AD-related proteins in NDAN donors.
Collapse
Affiliation(s)
- Batbayar Tumurbaatar
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, Texas, 77555 USA
| | - Anna Fracassi
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, Texas, 77555 USA
| | - Pietro Scaduto
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, Texas, 77555 USA
| | - Jutatip Guptarak
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, Texas, 77555 USA
| | - Randall Woltjer
- Department of Pathology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, USA
| | - Daniel Jupiter
- Department of Biostatistics & Data Science, Department of Orthopaedic Surgery and Rehabilitation, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, Texas, 77555 USA
| | - Giulio Taglialatela
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, Texas, 77555 USA
| |
Collapse
|
37
|
Lanctôt KL, Rivet L, Tumati S, Perin J, Sankhe K, Vieira D, Mintzer J, Rosenberg PB, Shade D, Lerner AJ, Padala PR, Brawman-Mintzer O, van Dyck CH, Porsteinsson AP, Craft S, Levey AI, Padala KP, Herrmann N. Heterogeneity of Response to Methylphenidate in Apathetic Patients in the ADMET 2 Trial. Am J Geriatr Psychiatry 2023; 31:1077-1087. [PMID: 37385898 PMCID: PMC10765607 DOI: 10.1016/j.jagp.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 07/01/2023]
Abstract
OBJECTIVE The Apathy in Dementia Methylphenidate Trial 2 (ADMET 2) found that methylphenidate was effective in treating apathy with a small-to-medium effect size but showed heterogeneity in response. We assessed clinical predictors of response to help determine individual likelihood of treatment benefit from methylphenidate. DESIGN Univariate and multivariate analyses of 22 clinical predictors of response chosen a priori. SETTING Data from the ADMET 2 randomized, placebo controlled multi-center clinical trial. PARTICIPANTS Alzheimer's disease patients with clinically significant apathy. MEASUREMENTS Apathy assessed with the Neuropsychiatric Inventory apathy domain (NPI-A). RESULTS In total, 177 participants (67% male, mean [SD] age 76.4 [7.9], mini-mental state examination 19.3 [4.8]) had 6-months follow up data. Six potential predictors met criteria for inclusion in multivariate modeling. Methylphenidate was more efficacious in participants without NPI anxiety (change in NPI-A -2.21, standard error [SE]:0.60) or agitation (-2.63, SE:0.68), prescribed cholinesterase inhibitors (ChEI) (-2.44, SE:0.62), between 52 and 72 years of age (-2.93, SE:1.05), had 73-80 mm Hg diastolic blood pressure (-2.43, SE: 1.03), and more functional impairment (-2.56, SE:1.16) as measured by the Alzheimer's Disease Cooperative Study Activities of Daily Living scale. CONCLUSION Individuals who were not anxious or agitated, younger, prescribed a ChEI, with optimal (73-80 mm Hg) diastolic blood pressure, or having more impaired function were more likely to benefit from methylphenidate compared to placebo. Clinicians may preferentially consider methylphenidate for apathetic AD participants already prescribed a ChEI and without baseline anxiety or agitation.
Collapse
Affiliation(s)
- Krista L Lanctôt
- Sunnybrook Research Institute (KLL, LR, ST, KS, DV, NH), University of Toronto, Toronto, Ontario, Canada.
| | - Luc Rivet
- Sunnybrook Research Institute (KLL, LR, ST, KS, DV, NH), University of Toronto, Toronto, Ontario, Canada
| | - Shankar Tumati
- Sunnybrook Research Institute (KLL, LR, ST, KS, DV, NH), University of Toronto, Toronto, Ontario, Canada
| | - Jamie Perin
- Bloomberg School of Public Health (JP, DS), Johns Hopkins University, Baltimore, MD
| | - Krushnaa Sankhe
- Sunnybrook Research Institute (KLL, LR, ST, KS, DV, NH), University of Toronto, Toronto, Ontario, Canada
| | - Danielle Vieira
- Sunnybrook Research Institute (KLL, LR, ST, KS, DV, NH), University of Toronto, Toronto, Ontario, Canada
| | - Jacobo Mintzer
- Ralph H. Johnson VA Medical Center (JM, OB-M), Medical University of South Carolina, Charleston, SC
| | - Paul B Rosenberg
- Department of Psychiatry and Behavioral Sciences (PBR), Johns Hopkins University, Baltimore, MD
| | - David Shade
- Bloomberg School of Public Health (JP, DS), Johns Hopkins University, Baltimore, MD
| | - Alan J Lerner
- University Hospitals Cleveland Medical Center (AJL), Case Western Reserve University School of Medicine, Cleveland, OH
| | - Prasad R Padala
- Central Arkansas Veterans Healthcare System (PRP, KPP), University of Arkansas for Medical Sciences, Little Rock, AR
| | - Olga Brawman-Mintzer
- Ralph H. Johnson VA Medical Center (JM, OB-M), Medical University of South Carolina, Charleston, SC
| | | | | | | | - Allan I Levey
- Emory Goizueta Alzheimer's Disease Research Center (AIL), Atlanta, GA
| | - Kalpana P Padala
- Central Arkansas Veterans Healthcare System (PRP, KPP), University of Arkansas for Medical Sciences, Little Rock, AR
| | - Nathan Herrmann
- Sunnybrook Research Institute (KLL, LR, ST, KS, DV, NH), University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
38
|
Namballa HK, Decker AM, Dorogan M, Gudipally A, Goclon J, Harding WW. Fluoroalkoxylated C-3 and C-9 (S)-12-bromostepholidine analogues with D1R antagonist activity. Bioorg Chem 2023; 141:106862. [PMID: 37722267 PMCID: PMC10872833 DOI: 10.1016/j.bioorg.2023.106862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/10/2023] [Accepted: 09/11/2023] [Indexed: 09/20/2023]
Abstract
To illuminate the tolerance of fluoroalkoxylated groups at the C-3 and C-9 positions of tetrahydroprotoberberines (THPBs) on D1R activity, C-3 and C-9 fluoroalkoxylated analogues of (S)-12-bromostepholidine were prepared and evaluated. All compounds examined were D1R antagonists as measured by a cAMP assay. Our structure-activity studies herein indicate that the C-3 position tolerates a 1,1-difluoroethoxy substituent for D1R antagonist activity. Compound 13a was the most potent cAMP-based D1R antagonist identified and was also found to antagonize β-arrestin translocation in a TANGO assay. Affinity assessments at other dopamine receptors revealed that 13a is selective for D1R and unlike other naturally-occurring THPBs such as (S)-stepholidine, lacks D2R affinity. In preliminary biopharmaceutical assays, excellent BBB permeation was observed for 13a. Further pharmacological studies are warranted on (S)-stepholidine congeners to harvest their potential as a source of novel, druggable D1R-targeted agents.
Collapse
Affiliation(s)
- Hari K Namballa
- Department of Chemistry, Hunter College, City University of New York, 695 Park Avenue, NY 10065, United States
| | - Ann M Decker
- Center for Drug Discovery, RTI International, Research Triangle Park, NC 27709, United States
| | - Michael Dorogan
- Department of Chemistry, Hunter College, City University of New York, 695 Park Avenue, NY 10065, United States
| | - Ashok Gudipally
- Department of Chemistry, Hunter College, City University of New York, 695 Park Avenue, NY 10065, United States; Program in Chemistry, CUNY Graduate Center 365 5th Avenue, New York, NY 10016, United States
| | - Jakub Goclon
- Department of Chemistry, Hunter College, City University of New York, 695 Park Avenue, NY 10065, United States
| | - Wayne W Harding
- Department of Chemistry, Hunter College, City University of New York, 695 Park Avenue, NY 10065, United States; Program in Chemistry, CUNY Graduate Center 365 5th Avenue, New York, NY 10016, United States; Program in Biochemistry, CUNY Graduate Center 365 5th Avenue, New York, NY 10016, United States.
| |
Collapse
|
39
|
Govindaraju R, Govindaraju S, Yun K, Kim J. Fluorescent-Based Neurotransmitter Sensors: Present and Future Perspectives. BIOSENSORS 2023; 13:1008. [PMID: 38131768 PMCID: PMC10742055 DOI: 10.3390/bios13121008] [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: 10/28/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023]
Abstract
Neurotransmitters (NTs) are endogenous low-molecular-weight chemical compounds that transmit synaptic signals in the central nervous system. These NTs play a crucial role in facilitating signal communication, motor control, and processes related to memory and learning. Abnormalities in the levels of NTs lead to chronic mental health disorders and heart diseases. Therefore, detecting imbalances in the levels of NTs is important for diagnosing early stages of diseases associated with NTs. Sensing technologies detect NTs rapidly, specifically, and selectively, overcoming the limitations of conventional diagnostic methods. In this review, we focus on the fluorescence-based biosensors that use nanomaterials such as metal clusters, carbon dots, and quantum dots. Additionally, we review biomaterial-based, including aptamer- and enzyme-based, and genetically encoded biosensors. Furthermore, we elaborate on the fluorescence mechanisms, including fluorescence resonance energy transfer, photon-induced electron transfer, intramolecular charge transfer, and excited-state intramolecular proton transfer, in the context of their applications for the detection of NTs. We also discuss the significance of NTs in human physiological functions, address the current challenges in designing fluorescence-based biosensors for the detection of NTs, and explore their future development.
Collapse
Affiliation(s)
- Rajapriya Govindaraju
- Department of Chemical and Biological Engineering, Gachon University, 1342 Seongnam Daero, Seongnam-si 13120, Gyeonggi-do, Republic of Korea;
| | - Saravanan Govindaraju
- Department of Bio Nanotechnology, Gachon University, Seongnam-si 13120, Gyeonggi-do, Republic of Korea; (S.G.); (K.Y.)
| | - Kyusik Yun
- Department of Bio Nanotechnology, Gachon University, Seongnam-si 13120, Gyeonggi-do, Republic of Korea; (S.G.); (K.Y.)
| | - Jongsung Kim
- Department of Chemical and Biological Engineering, Gachon University, 1342 Seongnam Daero, Seongnam-si 13120, Gyeonggi-do, Republic of Korea;
| |
Collapse
|
40
|
Stapleton MC, Koch SP, Cortes DRE, Wyman S, Schwab KE, Mueller S, McKennan CG, Boehm-Sturm P, Wu YL. Apolipoprotein-E deficiency leads to brain network alteration characterized by diffusion MRI and graph theory. Front Neurosci 2023; 17:1183312. [PMID: 38075287 PMCID: PMC10702609 DOI: 10.3389/fnins.2023.1183312] [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: 03/09/2023] [Accepted: 09/18/2023] [Indexed: 02/12/2024] Open
Abstract
Late-onset Alzheimer's disease (LOAD) is a major health concern for senior citizens, characterized by memory loss, confusion, and impaired cognitive abilities. Apolipoprotein-E (ApoE) is a well-known risk factor for LOAD, though exactly how ApoE affects LOAD risks is unknown. We hypothesize that ApoE attenuation of LOAD resiliency or vulnerability has a neurodevelopmental origin via changing brain network architecture. We investigated the brain network structure in adult ApoE knock out (ApoE KO) and wild-type (WT) mice with diffusion tensor imaging (DTI) followed by graph theory to delineate brain network topology. Left and right hemisphere connectivity revealed significant differences in number of connections between the hippocampus, amygdala, caudate putamen and other brain regions. Network topology based on the graph theory of ApoE KO demonstrated decreased functional integration, network efficiency, and network segregation between the hippocampus and amygdala and the rest of the brain, compared to those in WT counterparts. Our data show that brain network developed differently in ApoE KO and WT mice at 5 months of age, especially in the network reflected in the hippocampus, amygdala, and caudate putamen. This indicates that ApoE is involved in brain network development which might modulate LOAD risks via changing brain network structures.
Collapse
Affiliation(s)
- Margaret Caroline Stapleton
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Rangos Research Center Animal Imaging Core, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Stefan Paul Koch
- Charité 3R | Replace, Reduce, Refine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Experimental Neurology and Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Devin Raine Everaldo Cortes
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Rangos Research Center Animal Imaging Core, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Samuel Wyman
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Rangos Research Center Animal Imaging Core, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Kristina E. Schwab
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Rangos Research Center Animal Imaging Core, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Susanne Mueller
- Charité 3R | Replace, Reduce, Refine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Experimental Neurology and Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Philipp Boehm-Sturm
- Charité 3R | Replace, Reduce, Refine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Experimental Neurology and Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Yijen Lin Wu
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Rangos Research Center Animal Imaging Core, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| |
Collapse
|
41
|
Kabbej N, Ashby FJ, Riva A, Gamlin PD, Mandel RJ, Kunta A, Rouse CJ, Heldermon CD. Sex differences in brain transcriptomes of juvenile Cynomolgus macaques. RESEARCH SQUARE 2023:rs.3.rs-3422091. [PMID: 38045237 PMCID: PMC10690328 DOI: 10.21203/rs.3.rs-3422091/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Background: Behavioral, social, and physical characteristics are posited to distinguish the sexes, yet research on transcription-level sexual differences in the brain is limited. Here, we investigated sexually divergent brain transcriptomics in prepubertal cynomolgus macaques, a commonly used surrogate species to humans. Methods: A transcriptomic profile using RNA sequencing was generated for the temporal lobe, ventral midbrain, and cerebellum of 3 female and 3 male cynomolgus macaques previously treated with an Adeno-associated virus vector mix. Statistical analyses to determine differentially expressed protein-coding genes in all three lobes were conducted using DeSeq2 with a false discovery rate corrected P value of .05. Results: We identified target genes in the temporal lobe, ventral midbrain, and cerebellum with functions in translation, immunity, behavior, and neurological disorders that exhibited statistically significant sexually divergent expression. Conclusions: We provide potential mechanistic insights to the epidemiological differences observed between the sexes with regards to mental health and infectious diseases, such as COVID19. Our results provide pre-pubertal information on sexual differences in non-human primate brain transcriptomics and may provide insight to health disparities between the biological sexes in humans.
Collapse
|
42
|
Shaikh A, Ahmad F, Teoh SL, Kumar J, Yahaya MF. Targeting dopamine transporter to ameliorate cognitive deficits in Alzheimer's disease. Front Cell Neurosci 2023; 17:1292858. [PMID: 38026688 PMCID: PMC10679733 DOI: 10.3389/fncel.2023.1292858] [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: 09/12/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by the pathologic deposition of amyloid and neurofibrillary tangles in the brain, leading to neuronal damage and defective synapses. These changes manifest as abnormalities in cognition and behavior. The functional deficits are also attributed to abnormalities in multiple neurotransmitter systems contributing to neuronal dysfunction. One such important system is the dopaminergic system. It plays a crucial role in modulating movement, cognition, and behavior while connecting various brain areas and influencing other neurotransmitter systems, making it relevant in neurodegenerative disorders like AD and Parkinson's disease (PD). Considering its significance, the dopaminergic system has emerged as a promising target for alleviating movement and cognitive deficits in PD and AD, respectively. Extensive research has been conducted on dopaminergic neurons, receptors, and dopamine levels as critical factors in cognition and memory in AD. However, the exact nature of movement abnormalities and other features of extrapyramidal symptoms are not fully understood yet in AD. Recently, a previously overlooked element of the dopaminergic system, the dopamine transporter, has shown significant promise as a more effective target for enhancing cognition while addressing dopaminergic system dysfunction in AD.
Collapse
Affiliation(s)
- Ammara Shaikh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Fairus Ahmad
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Mohamad Fairuz Yahaya
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
43
|
Ismail H, Khalid D, Waseem D, Ijaz MU, Dilshad E, Haq IU, Bhatti MZ, Anwaar S, Ahmed M, Saleem S. Bioassays guided isolation of berberine from Berberis lycium and its neuroprotective role in aluminium chloride induced rat model of Alzheimer's disease combined with insilico molecular docking. PLoS One 2023; 18:e0286349. [PMID: 37910530 PMCID: PMC10619822 DOI: 10.1371/journal.pone.0286349] [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: 12/25/2022] [Accepted: 05/13/2023] [Indexed: 11/03/2023] Open
Abstract
OBJECTIVE Berberis lycium is an indigenous plant of Pakistan that is known for its medicinal properties. In the current study, we investigated the anti-Alzheimer's effect of berberine isolated from Berberis lycium. METHODS Root extract of B. lycium was subjected to acetylcholinesterase inhibition assay and column chromatography for bioassays guided isolation of a compound. The neuroprotective and memory improving effects of isolated compound were evaluated by aluminium chloride induced Alzheimer's disease rat model, elevated plus maze (EPM) and Morris water maze (MWM) tests., Levels of dopamine and serotonin in rats brains were determined using HPLC. Moreover, western blot and docking were performed to determine interaction between berberine and β-secretase. RESULTS During fractionation, ethyl acetate and methanol (3:7) fraction was collected from solvent mixture of ethyl acetate and methanol. This fraction showed the highest anti-acetylcholinesterase activity and was alkaloid positive. The results of TLC and HPLC analysis indicated the presence of the isolated compound as berberine. Additionally, the confirmation of isolated compound as berberine was carried out using FTIR and NMR analysis. In vivo EPM and MWM tests showed improved memory patterns after berberine treatment in Alzheimer's disease model. The levels of dopamine, serotonin and activity of antioxidant enzymes were significantly (p<0.05) enhanced in brain tissue homogenates of berberine treated group. This was supported by decreased expression of β-secretase in berberine treated rat brain homogenates and good binding affinity of berberine with β-secretase in docking studies. Binding energies for interaction of β-secretase with berberine and drug Rivastigmine is -7.0 kcal/mol and -5.8 kcal/mol respectively representing the strong interactions. The results of docked complex of secretase with berberine and Rivastigmine was carried out using Gromacs which showed significant stability of complex in terms of RMSD and radius of gyration. Overall, the study presents berberine as a potential drug against Alzheimer's disease by providing evidence of its effects in improving memory, neurotransmitter levels and reducing β-secretase expression in the Alzheimer's disease model.
Collapse
Affiliation(s)
- Hammad Ismail
- Department of Biochemistry and Biotechnology, University of Gujrat, Gujrat, Pakistan
| | - Dania Khalid
- Department of Biochemistry and Biotechnology, University of Gujrat, Gujrat, Pakistan
| | - Durdana Waseem
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Muhammad Umar Ijaz
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan
| | - Erum Dilshad
- Department of Bioinformatics and Biosciences, Faculty of Health and Life Sciences, Capital University of Science and Technology, Islamabad, Pakistan
| | - Ihsan-ul Haq
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| | - Muhammad Zeeshan Bhatti
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| | - Sadaf Anwaar
- Department of Biological Sciences, International Islamic University, Islamabad, Pakistan
| | - Madiha Ahmed
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Samreen Saleem
- Department of Nutrition and Lifestyle Medicine, Health Services Academy, Islamabad, Pakistan
| |
Collapse
|
44
|
Tian J, Du E, Guo L. Mitochondrial Interaction with Serotonin in Neurobiology and Its Implication in Alzheimer's Disease. J Alzheimers Dis Rep 2023; 7:1165-1177. [PMID: 38025801 PMCID: PMC10657725 DOI: 10.3233/adr-230070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 08/16/2023] [Indexed: 12/01/2023] Open
Abstract
Alzheimer's disease (AD) is a lethal neurodegenerative disorder characterized by severe brain pathologies and progressive cognitive decline. While the exact cause of this disease remains unknown, emerging evidence suggests that dysregulation of neurotransmitters contributes to the development of AD pathology and symptoms. Serotonin, a critical neurotransmitter in the brain, plays a pivotal role in regulating various brain processes and is implicated in neurological and psychiatric disorders, including AD. Recent studies have shed light on the interplay between mitochondrial function and serotonin regulation in brain physiology. In AD, there is a deficiency of serotonin, along with impairments in mitochondrial function, particularly in serotoninergic neurons. Additionally, altered activity of mitochondrial enzymes, such as monoamine oxidase, may contribute to serotonin dysregulation in AD. Understanding the intricate relationship between mitochondria and serotonin provides valuable insights into the underlying mechanisms of AD and identifies potential therapeutic targets to restore serotonin homeostasis and alleviate AD symptoms. This review summarizes the recent advancements in unraveling the connection between brain mitochondria and serotonin, emphasizing their significance in AD pathogenesis and underscoring the importance of further research in this area. Elucidating the role of mitochondria in serotonin dysfunction will promote the development of therapeutic strategies for the treatment and prevention of this neurodegenerative disorder.
Collapse
Affiliation(s)
- Jing Tian
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| | - Eric Du
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
- Blue Valley West High School, Overland Park, KS, USA
| | - Lan Guo
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
45
|
Luo Y, Yang FY, Lo RY. Application of transcranial brain stimulation in dementia. Tzu Chi Med J 2023; 35:300-305. [PMID: 38035058 PMCID: PMC10683520 DOI: 10.4103/tcmj.tcmj_91_23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/19/2023] [Accepted: 06/01/2023] [Indexed: 12/02/2023] Open
Abstract
The number of patients with dementia grows rapidly as the global population ages, which posits tremendous health-care burden to the society. Only cholinesterase inhibitors and a N-methyl-D-aspartate receptor antagonist have been approved for treating patients with Alzheimer's disease (AD), and their clinical effects remained limited. Medical devices serve as an alternative therapeutic approach to modulating neural activities and enhancing cognitive function. Four major brain stimulation technologies including deep brain stimulation (DBS), transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), and transcranial ultrasound stimulation (TUS) have been applied to AD in a clinical trial setting. DBS allows electrical stimulation at the specified nucleus but remains resource-demanding, and after all, an invasive surgery; whereas TMS and tDCS are widely available and affordable but less ideal with respect to localization. The unique physical property of TUS, on the other hand, allows both thermal and mechanical energy to be transduced and focused for neuromodulation. In the context of dementia, using focused ultrasound to induce blood-brain barrier opening for delivering drugs and metabolizing amyloid protein has drawn great attention in recent years. Furthermore, low-intensity pulsed ultrasound has demonstrated its neuroprotective effects in both in vitro and in vivo studies, leading to ongoing clinical trials for AD. The potential and limitation of transcranial brain stimulation for treating patients with dementia would be discussed in this review.
Collapse
Affiliation(s)
- Yuncin Luo
- Department of Neurology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation and Tzu Chi University, Hualien, Taiwan
| | - Feng-Yi Yang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Raymond Y. Lo
- Department of Neurology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation and Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
46
|
Berg SZ, Berg J. Melanin: a unifying theory of disease as exemplified by Parkinson's, Alzheimer's, and Lewy body dementia. Front Immunol 2023; 14:1228530. [PMID: 37841274 PMCID: PMC10570809 DOI: 10.3389/fimmu.2023.1228530] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/13/2023] [Indexed: 10/17/2023] Open
Abstract
Melanin, a ubiquitous dark pigment, plays important roles in the immune system, including scavenging reactive oxygen species formed in response to ultraviolet radiation absorption, absorbing metals, thermal regulation, drug uptake, innate immune system functions, redox, and energy transduction. Many tissue types, including brain, heart, arteries, ovaries, and others, contain melanin. Almost all cells contain precursors to melanin. A growing number of diseases in which there is a loss of melanin and/or neuromelanin are increasingly thought to have infectious etiologies, for example, Alzheimer's disease (AD), Parkinson's disease (PD), Lewy Body Dementia (LBD), and vitiligo. AD, PD, LBD, and vitiligo have been linked with herpesvirus, which enters melanosomes and causes apoptosis, and with gut dysbiosis and inflammation. Herpesvirus is also linked with gut dysbiosis and inflammation. We theorize that under normal healthy states, melanin retains some of the energy it absorbs from electromagnetic radiation, which is then used to fuel cells, and energy from ATP is used to compliment that energy supply. We further theorize that loss of melanin reduces the energy supply of cells, which in the case of AD, PD, and LBD results in an inability to sustain immune system defenses and remove the plaques associated with the disease, which appear to be part of the immune system's attempt to eradicate the pathogens seen in these neurodegenerative diseases. In addition, in an attempt to explain why removing these plaques does not result in improvements in cognition and mood and why cognitions and moods in these individuals have ebbs and flows, we postulate that it is not the plaques that cause the cognitive symptoms but, rather, inflammation in the brain resulting from the immune system's response to pathogens. Our theory that energy retained in melanin fuels cells in an inverse relationship with ATP is supported by studies showing alterations in ATP production in relationship to melanin levels in melanomas, vitiligo, and healthy cells. Therefore, alteration of melanin levels may be at the core of many diseases. We propose regulating melanin levels may offer new avenues for treatment development.
Collapse
Affiliation(s)
- Stacie Z. Berg
- Department of Translational Biology, William Edwards LLC, Baltimore, MD, United States
| | - Jonathan Berg
- Department of Translational Biology, William Edwards LLC, Baltimore, MD, United States
| |
Collapse
|
47
|
Abstract
Because the central nervous system is largely nonrenewing, neurons and their synapses must be maintained over the lifetime of an individual to ensure circuit function. Age is a dominant risk factor for neural diseases, and declines in nervous system function are a common feature of aging even in the absence of disease. These alterations extend to the visual system and, in particular, to the retina. The retina is a site of clinically relevant age-related alterations but has also proven to be a uniquely approachable system for discovering principles that govern neural aging because it is well mapped, contains diverse neuron types, and is experimentally accessible. In this article, we review the structural and molecular impacts of aging on neurons within the inner and outer retina circuits. We further discuss the contribution of non-neuronal cell types and systems to retinal aging outcomes. Understanding how and why the retina ages is critical to efforts aimed at preventing age-related neural decline and restoring neural function.
Collapse
Affiliation(s)
- Jeffrey D Zhu
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA;
| | - Sharma Pooja Tarachand
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA;
| | - Qudrat Abdulwahab
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA;
| | - Melanie A Samuel
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA;
| |
Collapse
|
48
|
Salgueiro WG, Soares MV, Martins CF, Paula FR, Rios-Anjos RM, Carrazoni T, Mori MA, Müller RU, Aschner M, Dal Belo CA, Ávila DS. Dopaminergic modulation by quercetin: In silico and in vivo evidence using Caenorhabditis elegans as a model. Chem Biol Interact 2023; 382:110610. [PMID: 37348670 PMCID: PMC10527449 DOI: 10.1016/j.cbi.2023.110610] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 06/12/2023] [Accepted: 06/19/2023] [Indexed: 06/24/2023]
Abstract
Quercetin is a flavonol widely distributed in plants and has various described biological functions. Several studies have reported on its ability to restore neuronal function in a wide variety of disease models, including animal models of neurodegenerative disorders such as Parkinson's disease. Quercetin per se can act as a neuroprotector/neuromodulator, especially in diseases related to impaired dopaminergic neurotransmission. However, little is known about how quercetin interacts with the dopaminergic machinery. Here we employed the nematode Caenorhabditis elegans to study this putative interaction. After observing behavioral modulation, mutant analysis and gene expression in C. elegans upon exposure to quercetin at a concentration that does not protect against MPTP, we constructed a homology-based dopamine transporter protein model to conduct a docking study. This led to suggestive evidence on how quercetin may act as a dopaminergic modulator by interacting with C. elegans' dopamine transporter and alter the nematode's exploratory behavior. Consistent with this model, quercetin controls C. elegans behavior in a way dependent on the presence of both the dopamine transporter (dat-1), which is up-regulated upon quercetin exposure, and the dopamine receptor 2 (dop-2), which appears to be mandatory for dat-1 up-regulation. Our data propose an interaction with the dopaminergic machinery that may help to establish the effects of quercetin as a neuromodulator.
Collapse
Affiliation(s)
- Willian Goulart Salgueiro
- Research Group in Biochemistry and Toxicology in Caenorhabditis elegans (GBToxCe), Federal University of Pampa - UNIPAMPA, CEP 97500-970, Uruguaiana, RS, Brazil; Department of Biochemistry and Tissue Biology, University of Campinas, Monteiro Lobato Avenue, 255, Campinas, São Paulo, 13083-862, Brazil; Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany; Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Marcell Valandro Soares
- Research Group in Biochemistry and Toxicology in Caenorhabditis elegans (GBToxCe), Federal University of Pampa - UNIPAMPA, CEP 97500-970, Uruguaiana, RS, Brazil; Department of Biochemistry and Molecular Biology, Post-graduate Program in Biological Sciences, Federal University of Santa Maria, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Cassiano Fiad Martins
- Research Group in Biochemistry and Toxicology in Caenorhabditis elegans (GBToxCe), Federal University of Pampa - UNIPAMPA, CEP 97500-970, Uruguaiana, RS, Brazil
| | - Fávero Reisdorfer Paula
- Laboratory for Development and Quality Control in Medicines (LDCQ), Federal University of Pampa, Uruguaiana, RS, Brazil
| | | | - Thiago Carrazoni
- Neurobiology and Toxinology Laboratory, (LANETOX), Federal University of Pampa - UNIPAMPA, CEP 97300-000, São Gabriel, RS, Brazil
| | - Marcelo A Mori
- Department of Biochemistry and Tissue Biology, University of Campinas, Monteiro Lobato Avenue, 255, Campinas, São Paulo, 13083-862, Brazil; Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, SP, Brazil; Experimental Medicine Research Cluster, University of Campinas, Campinas, SP, Brazil
| | - Roman-Ulrich Müller
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany; Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Cháriston André Dal Belo
- Department of Biochemistry and Molecular Biology, Post-graduate Program in Biological Sciences, Federal University of Santa Maria, Camobi, 97105-900, Santa Maria, RS, Brazil; Neurobiology and Toxinology Laboratory, (LANETOX), Federal University of Pampa - UNIPAMPA, CEP 97300-000, São Gabriel, RS, Brazil; Multidisciplinar Department, Federal University of São Paulo (UNIFESP), Angelica Street, 100- CEP 06110295, Osasco, SP, Brazil
| | - Daiana Silva Ávila
- Research Group in Biochemistry and Toxicology in Caenorhabditis elegans (GBToxCe), Federal University of Pampa - UNIPAMPA, CEP 97500-970, Uruguaiana, RS, Brazil; Department of Biochemistry and Molecular Biology, Post-graduate Program in Biological Sciences, Federal University of Santa Maria, Camobi, 97105-900, Santa Maria, RS, Brazil.
| |
Collapse
|
49
|
Pilozzi A, Foster S, Mischoulon D, Fava M, Huang X. A Brief Review on the Potential of Psychedelics for Treating Alzheimer's Disease and Related Depression. Int J Mol Sci 2023; 24:12513. [PMID: 37569888 PMCID: PMC10419627 DOI: 10.3390/ijms241512513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 08/03/2023] [Accepted: 08/05/2023] [Indexed: 08/13/2023] Open
Abstract
Alzheimer's disease (AD), the most common form of senile dementia, is poised to place an even greater societal and healthcare burden as the population ages. With few treatment options for the symptomatic relief of the disease and its unknown etiopathology, more research into AD is urgently needed. Psychedelic drugs target AD-related psychological pathology and symptoms such as depression. Using microdosing, psychedelic drugs may prove to help combat this devastating disease by eliciting psychiatric benefits via acting through various mechanisms of action such as serotonin and dopamine pathways. Herein, we review the studied benefits of a few psychedelic compounds that may show promise in treating AD and attenuating its related depressive symptoms. We used the listed keywords to search through PubMed for relevant preclinical, clinical research, and review articles. The putative mechanism of action (MOA) for psychedelics is that they act mainly as serotonin receptor agonists and induce potential beneficial effects for treating AD and related depression.
Collapse
Affiliation(s)
- Alexander Pilozzi
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Simmie Foster
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
- Depression Clinical & Research Program, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - David Mischoulon
- Depression Clinical & Research Program, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Maurizio Fava
- Depression Clinical & Research Program, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Xudong Huang
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
50
|
Hawash ZAS, Yassien EM, Alotaibi BS, El-Moslemany AM, Shukry M. Assessment of Anti-Alzheimer Pursuit of Jambolan Fruit Extract and/or Choline against AlCl 3 Toxicity in Rats. TOXICS 2023; 11:509. [PMID: 37368609 DOI: 10.3390/toxics11060509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/30/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023]
Abstract
Jambolan fruit extract and choline were investigated for Aluminum tri chloride (AlCl3)-induced Alzheimer's disease in rats. Thirty-six male "Sprague Dawley" rats weighing (150 ± 10 g) were allocated into six groups; the first group was fed a baseline diet and served as a negative control. Alzheimer's disease (AD) was induced in Group 2 rats by oral administration of AlCl3 (17 mg/kg body weight) dissolved in distilled water (served as a positive control). Rats in Group 3 were orally supplemented concomitantly with both 500 mg/kg BW of an ethanolic extract of jambolan fruit once daily for 28 days and AlCl3 (17 mg/kg body weight). Group 4: Rivastigmine (RIVA) aqueous infusion (0.3 mg/kg BW/day) was given orally to rats as a reference drug concomitantly with oral supplementation of AlCl3 (17 mg/kg body weight) for 28 days. Group 5 rats were orally treated with choline (1.1 g/kg) concomitantly with oral supplementation of AlCl3 (17 mg/kg body weight). Group 6 was given 500 mg/kg of jambolan fruit ethanolic extract and 1.1 g/kg of choline orally to test for additive effects concurrently with oral supplementation of AlCl3 (17 mg/kg bw) for 28 days. Body weight gain, feed intake, feed efficiency ratio, and relative brain, liver, kidney, and spleen weight were calculated after the trial. Brain tissue assessment was analyzed for antioxidant/oxidant markers, biochemical analysis in blood serum, a phenolic compound in Jambolan fruits extracted by high-performance liquid chromatography (HPLC), and histopathology of the brain. The results showed that Jambolan fruit extract and choline chloride improved brain functions, histopathology, and antioxidant enzyme activity compared with the positive group. In conclusion, administering jambolan fruit extract and choline can lower the toxic impacts of aluminum chloride on the brain.
Collapse
Affiliation(s)
- Zeinab Abdel Salam Hawash
- Nutrition and Food Science Department, Faculty of Home Economic, Al-Azhar University, Tanta 31732, Egypt
| | - Ensaf M Yassien
- Nutrition and Food Science Department, Faculty of Home Economic, Al-Azhar University, Tanta 31732, Egypt
| | - Badriyah S Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Amira M El-Moslemany
- Nutrition and Food Science Department, Faculty of Home Economic, Al-Azhar University, Tanta 31732, Egypt
| | - Mustafa Shukry
- Physiology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| |
Collapse
|