1
|
Serebrovska Z, Xi L, Fedoriuk M, Dosenko V, Shysh A, Khetsuriani M, Porkhalo D, Savchenko A, Goncharov S, Utko N, Virko S, Kholin V, Egorov E, Koval R, Maksymchuk O. Intermittent hypoxia-hyperoxia training ameliorates cognitive impairment and neuroinflammation in a rat model of Alzheimer's disease. Brain Res 2025; 1847:149301. [PMID: 39476996 DOI: 10.1016/j.brainres.2024.149301] [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: 02/22/2024] [Revised: 10/02/2024] [Accepted: 10/26/2024] [Indexed: 11/15/2024]
Abstract
Alzheimer's disease (AD), characterized by severe and progressive cognitive decline, stands as one of the most prevalent and devastating forms of dementia. Based on our recent findings showing intermittent hypoxic conditioning improved neuronal function in patients with mild cognitive impairment, the present study aimed at investigating whether the neuroprotective effects of intermittent hypoxia can be replicated in a rat model of AD, which allows us to explore the underlying cellular mechanisms involving neuroinflammation, hypoxia inducible factor 1α (HIF1α), and cytochrome P450 family 2 subfamily E member 1 (CYP2E1). Forty-one adult male Wistar rats were randomly assigned to three groups: 1) Control group: received intracerebroventricular (ICV) injection of saline; 2) STZ group: received ICV injection of streptozotocin (STZ) to induce AD-like pathology; and 3) STZ + IHHT group received ICV injection of STZ as well as 15 daily sessions of intermittent hypoxia-hyperoxia training (IHHT). We observed that ICV injection of STZ inhibited spatial learning and memory in the rats assessed with Morris Water Maze test. The cognitive function declines were accompanied by increased expression of amyloid β peptide (Aβ), HIF1α, CYP2E1, and TNFα in hippocampus. Interestingly, IHHT significantly restored the STZ-induced cognitive dysfunction, while reduced expression of Aβ, CYP2E1, HIF1α and TNFα. We conclude that IHHT with mild hypoxia-hyperoxia can enhance spatial learning and memory and reduce the AD-like pathologic changes in rats. The neuroprotective outcome of IHHT may be related to anti-inflammatory effects in hippocampus.
Collapse
Affiliation(s)
- Zoya Serebrovska
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv 01601, Ukraine.
| | - Lei Xi
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298-0204, USA.
| | - Mykhailo Fedoriuk
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv 01601, Ukraine
| | - Victor Dosenko
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv 01601, Ukraine
| | - Angela Shysh
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv 01601, Ukraine
| | - Michael Khetsuriani
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv 01601, Ukraine
| | - Denys Porkhalo
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv 01601, Ukraine
| | - Anton Savchenko
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv 01601, Ukraine
| | - Serhii Goncharov
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv 01601, Ukraine
| | - Natalie Utko
- Chebotarev Institute of Gerontology, National Academy of Sciences of Ukraine, Kyiv 04114, Ukraine
| | - Sergii Virko
- Lashkariov Institute of Semiconductor Physics, National Academy of Sciences, Kyiv 02000, Ukraine
| | - Victor Kholin
- Chebotarev Institute of Gerontology, National Academy of Sciences of Ukraine, Kyiv 04114, Ukraine
| | - Egor Egorov
- Institute for Preventive and Antiaging Medicine, Berlin 10789, Germany
| | - Roman Koval
- National Cancer Institute, Kyiv 03022, Ukraine
| | - Oksana Maksymchuk
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv 03143, Ukraine
| |
Collapse
|
2
|
Smith GC, Griffith KR, Sicher AR, Brockway DF, Proctor EA, Crowley NA. Alcohol consumption confers lasting impacts on prefrontal cortical neuron intrinsic excitability and spontaneous neurotransmitter signaling in the aging brain in mice. Neurobiol Aging 2025; 145:42-54. [PMID: 39476434 DOI: 10.1016/j.neurobiolaging.2024.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 11/06/2024]
Abstract
Both alcohol use disorder (AUD) and cognitive decline include disruption in the balance of excitation and inhibition in the cortex, but the potential role of alcohol use on excitation and inhibition on the aging brain is unclear. We examined the effect of moderate voluntary binge alcohol consumption on the aged, pre-disease neuronal environment by measuring intrinsic excitability and spontaneous neurotransmission on prefrontal cortical pyramidal (excitatory, glutamatergic) and non-pyramidal (inhibitory, GABAergic) neurons following a prolonged period of abstinence from alcohol in mice. Results highlight that binge alcohol consumption has lasting impacts on the electrophysiological properties of prefrontal cortical neurons. A profound increase in excitatory events onto layer 2/3 non-pyramidal neurons following alcohol consumption was seen, along with altered intrinsic excitability of pyramidal neurons, which could have a range of effects on cognitive disorder progression, such as Alzheimer's Disease, in humans. These results indicate that moderate voluntary alcohol influences the pre-disease environment in aging and highlight the need for further mechanistic investigation into this risk factor.
Collapse
Affiliation(s)
- Grace C Smith
- Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA; Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA; Penn State Neuroscience Institute, University Park, PA 16802, USA
| | - Keith R Griffith
- Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA; Penn State Neuroscience Institute, University Park, PA 16802, USA
| | - Avery R Sicher
- Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA; Neuroscience Graduate Program, Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA; Penn State Neuroscience Institute, University Park, PA 16802, USA
| | - Dakota F Brockway
- Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA; Neuroscience Graduate Program, Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA; Penn State Neuroscience Institute, University Park, PA 16802, USA
| | - Elizabeth A Proctor
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA; Department of Engineering Science and Mechanics, University Park, PA 16802, USA; Departments of Neurosurgery Penn State College of Medicine, University Park, PA 16802, USA; Penn State Neuroscience Institute, University Park, PA 16802, USA; Departments of Neurosurgery Penn State College of Medicine, Hershey, PA 17033, USA
| | - Nicole A Crowley
- Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA; Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA; Departments of Neurosurgery Penn State College of Medicine, University Park, PA 16802, USA; Penn State Neuroscience Institute, University Park, PA 16802, USA.
| |
Collapse
|
3
|
Padhan P, Simran, Kumar N, Verma S. Glutathione S-transferase: A keystone in Parkinson's disease pathogenesis and therapy. Mol Cell Neurosci 2024; 132:103981. [PMID: 39644945 DOI: 10.1016/j.mcn.2024.103981] [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: 09/26/2024] [Revised: 11/01/2024] [Accepted: 11/30/2024] [Indexed: 12/09/2024] Open
Abstract
Parkinson's disease is a progressive neurodegenerative disorder that predominantly affects motor function due to the loss of dopaminergic neurons in the substantia nigra. It presents significant challenges, impacting millions worldwide with symptoms such as tremors, rigidity, bradykinesia, and postural instability, leading to decreased quality of life and increased morbidity. The pathogenesis of Parkinson's disease is multifaceted, involving complex interactions between genetic susceptibility, environmental factors, and aging, with oxidative stress playing a central role in neuronal degeneration. Glutathione S-Transferase enzymes are critical in the cellular defense mechanism against oxidative stress, catalysing the conjugation of the antioxidant glutathione to various toxic compounds, thereby facilitating their detoxification. Recent research underscores the importance of Glutathione S-Transferase in the pathophysiology of Parkinson's disease, revealing that genetic polymorphisms in Glutathione S-Transferase genes influence the risk and progression of the disease. These genetic variations can affect the enzymatic activity of Glutathione S-Transferase, thereby modulating an individual's capacity to detoxify reactive oxygen species and xenobiotics, which are implicated in Parkinson's disease neuropathological processes. Moreover, biochemical studies have elucidated the role of Glutathione S-Transferase in not only maintaining cellular redox balance but also in modulating various cellular signalling pathways, highlighting its neuroprotective potential. From a therapeutic perspective, targeting Glutathione S-Transferase pathways offers promising avenues for the development of novel treatments aimed at enhancing neuroprotection and mitigating disease progression. This review explores the evident and hypothesized roles of Glutathione S-Transferase in Parkinson's disease, providing a comprehensive overview of its importance and potential as a target for therapeutic intervention.
Collapse
Affiliation(s)
- Pratyush Padhan
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Simran
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Neeraj Kumar
- Department of Reproductive Biology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Sonia Verma
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
4
|
Feng R, Zhu Q, Wang A, Wang H, Wang J, Chen P, Zhang R, Liang D, Teng J, Ma M, Ding X, Wang X. Effect of fecal microbiota transplantation on patients with sporadic amyotrophic lateral sclerosis: a randomized, double-blind, placebo-controlled trial. BMC Med 2024; 22:566. [PMID: 39617896 PMCID: PMC11610222 DOI: 10.1186/s12916-024-03781-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disorder marked by the progressive loss of motor neurons. Recent insights into ALS pathogenesis underscore the pivotal role of the gut microbiome, prompting an investigation into the potential therapeutic impact of fecal microbiota transplantation (FMT) on sporadic ALS patients. METHODS Conducted as a double-blind, placebo-controlled, parallel-group, randomized clinical trial, the study enrolled 27 participants from October 2022 to April 2023. The participants were followed up for 6 months from February 2023 to October 2023, during in-person visits at baseline, week 15, week 23, and week 35. The participants, evenly randomized, received either healthy donor FMT (FMT, n = 14) or a mixture of 0.9% saline and food coloring (E150c) as sham transplantation (placebo, n = 13). The primary outcome measured the change in the ALS Functional Rating Scale-Revised (ALSFRS-R) total score from baseline to week 35. Secondary outcomes included changes in gastrointestinal and respiratory functions, muscle strength, autonomic function, cognition, quality of life, intestinal microbiome composition, and plasm neurofilament light chain protein (NFL). Efficacy and safety outcomes were assessed in the intention-to-treat population. RESULTS A total of 27 randomized patients (47% women; mean age, 67.2 years), 24 participants completed the entire study. Notably, ALSFRS-R score changes exhibited no significant differences between FMT (6.1 [SD, 3.11]) and placebo (6.41[SD, 2.73]) groups from baseline to week 35. Secondary efficacy outcomes, encompassing respiratory function, muscle strength, autonomic function, cognition, quality of life, and plasm NFL, showed no significant differences. Nevertheless, the FMT group exhibited improvements in constipation, depression, and anxiety symptoms. FMT induced a shift in gut microbiome community composition, marked by increased abundance of Bifidobacterium, which persisted until week 15 (95% CI, 0.04 to 0.28; p = 0.01). Gastrointestinal adverse events were the primary manifestations of FMT-related side effects. CONCLUSIONS In this clinical trial involving 27 sporadic ALS patients, FMT did not significantly slow the decline in ALSFRS-R score. Larger multicenter trials are needed to confirm the efficacy of FMT in sporadic ALS patients and to explore the underlying biological mechanisms. TRIAL REGISTRATION Chinese Clinical Trial Registry Identifier: ChiCTR 2200064504.
Collapse
Affiliation(s)
- Renyi Feng
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, Henan, China
| | - Qingyong Zhu
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, Henan, China
| | - Ao Wang
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, Henan, China
| | - Hanzhen Wang
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, Henan, China
| | - Jiuqi Wang
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, Henan, China
| | - Pei Chen
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, Henan, China
| | - Rui Zhang
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, Henan, China
| | - Dongxiao Liang
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, Henan, China
| | - Junfang Teng
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, Henan, China
| | - Mingming Ma
- Department of Neurology, Affiliated People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China.
| | - Xuebing Ding
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China.
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, Henan, China.
| | - Xuejing Wang
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China.
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, Henan, China.
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China.
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
5
|
Chavez CLJ, Scheldrup GP, Madory LE, Denning CJE, Lee EC, Nguyen DT, Castro M, Garcia A, Torres‐Gonzales J, Herbert JN, Kotlyar D, Riazat N, Pakter W, Le W, Van Doren E, Ter Galstian M, Szumlinski KK. Biochemical changes precede affective and cognitive anomalies in aging adult C57BL/6J mice with a prior history of adolescent alcohol binge-drinking. Addict Biol 2024; 29:e70006. [PMID: 39665499 PMCID: PMC11635696 DOI: 10.1111/adb.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 12/13/2024]
Abstract
The early initiation of binge-drinking and biological sex are critical risk factors for the development of affective disturbances and cognitive decline, as well as neurodegenerative diseases including Alzheimer's disease. Further, a history of excessive alcohol consumption alters normal age-related changes in the pattern of protein expression in the brain, which may relate to an acceleration of cognitive decline. Here, we aimed to disentangle the interrelation between a history of binge-drinking during adolescence, biological sex and normal aging on the manifestation of negative affect, cognitive decline and associated biochemical pathology. To this end, adolescent male and female C57BL/6J mice (PND 28-29) underwent 30 days of alcohol binge-drinking using a modified drinking-in-the-dark (DID) paradigm. Then, mice were assayed for negative affect, sensorimotor gating and cognition at three developmental stages during adulthood-mature adulthood (6 months), pre-middle age (9 months) and middle age (12 months). Behavioural testing was then followed by immunoblotting to index the protein expression of glutamate receptors, neuropathological markers [Tau, p (Thr217)-Tau, p (Ser396)-Tau, BACE, APP, Aβ], as well as ERK activation within the entorhinal cortex, prefrontal cortex and amygdala. Across this age span, we detected only a few age-related changes in our measures of negative affect or spatial learning/memory in the Morris water maze and all of these changes were sex-specific. Prior adolescent binge-drinking impaired behaviour only during reversal learning in 9-month-old females and during radial arm maze testing in 12-month-old females. In contrast to behaviour, we detected a large number of protein changes related to prior binge-drinking history, several of which manifested as early as 6 months of age, with the prefrontal cortex particularly affected at this earlier age. While 6-month-old mice exhibited relatively few alcohol-related protein changes within the entorhinal cortex and amygdala, the number of alcohol-related protein changes within the entorhinal cortex increased with age, while the 12-month-old mice exhibited the largest number of protein changes within the amygdala. Approximately a third of the alcohol-related protein changes were sex-selective. Taken together, the results of our longitudinal study using a murine model of binge-drinking indicate that a prior history of heavy alcohol consumption, beginning in adolescence, is sufficient to induce what we presume to be latent changes in protein indices of cellular activity, glutamate transmission and neuropathology within key brain regions governing cognition, executive function and emotion that appear to precede the onset of robust behavioural signs of dysregulated affect and cognitive impairment.
Collapse
Affiliation(s)
- C. Leonardo Jimenez Chavez
- Department of Psychological and Brain SciencesUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Gavin P. Scheldrup
- Department of Psychological and Brain SciencesUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Lauren E. Madory
- Department of Psychological and Brain SciencesUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Christopher J. E. Denning
- Department of Psychological and Brain SciencesUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Edward C. Lee
- Department of Psychological and Brain SciencesUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Dylan T. Nguyen
- Department of Molecular, Cellular and Developmental BiologyUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Marian Castro
- Department of Psychological and Brain SciencesUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Andrew Garcia
- Department of Psychological and Brain SciencesUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Jose Torres‐Gonzales
- Department of Psychological and Brain SciencesUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Jessica N. Herbert
- Department of Psychological and Brain SciencesUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Daniel Kotlyar
- Department of Psychological and Brain SciencesUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Neda Riazat
- Department of Psychological and Brain SciencesUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| | - William Pakter
- Department of Psychological and Brain SciencesUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| | - William Le
- Department of Psychological and Brain SciencesUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Eliyanna Van Doren
- Department of Psychological and Brain SciencesUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Marianna Ter Galstian
- Department of Psychological and Brain SciencesUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Karen K. Szumlinski
- Department of Psychological and Brain SciencesUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
- Department of Molecular, Cellular and Developmental BiologyUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
- Neuroscience Research InstituteUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
- Center for Aging and Longevity StudiesUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| |
Collapse
|
6
|
Tian M, Zhang B, Liu H, Han Y, Yang M, Duan R, Guo D, Song C, Ma J, Liu Y. The Effect of Alcohol Consumption on Progressive Supranuclear Palsy: A Cross-Sectional Study. CNS Neurosci Ther 2024; 30:e70146. [PMID: 39713959 DOI: 10.1111/cns.70146] [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: 07/15/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 12/24/2024] Open
Abstract
AIMS To investigate the effect of alcohol consumption on the clinical symptoms in a cohort of Progressive supranuclear palsy (PSP) patients. METHODS We conducted a cross-sectional study focusing on possible and probable PSP patients in Qilu Hospital of Shandong University. Diagnoses and clinical phenotypes were confirmed using the 2017 Movement Disorder Society criteria and the Multiple Allocations eXtinction (MAX) rules. Data on drinking habits and demographics were collected via face-to-face interviews and medical records reviews. Clinical scales assessed motor and nonmotor symptoms. Alcohol consumption was categorized into light, moderate, and heavy status. Using multivariate linear regression and adjusting for confounding factors, we analyzed the relationship between alcohol consumption and clinical symptoms. RESULTS The study comprised 128 participants (59.4% male and 45.31% drinkers). Alcohol consumption has been associated with severe PSP clinical symptoms, particularly among male patients. Compared with nondrinkers, consumers of alcohol exhibit significantly more severe motor symptoms and cognitive impairments, particularly in the domains of visuospatial and executive abilities, memory, and language. Moreover, when categorizing individuals based on their intake of alcohol weekly, those with heavy consumption show significantly higher PSP Rating Scale (PSPRS) and the Unified Parkinson's Disease Rating Scale (UPDRS) scores, as well as significantly lower Montreal Cognitive Assessment Scale (MoCA) and Mini Mental State Examination (MMSE) scores compared to nonconsumers. CONCLUSION Our findings indicate an association between heavy alcohol consumption and more pronounced symptoms of PSP, especially cognitive function. It raises the possibility that alcohol intake may play a role in modulating the clinical course of PSP.
Collapse
Affiliation(s)
- Min Tian
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Bohan Zhang
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Heyin Liu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Yinlian Han
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Mu Yang
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Ruonan Duan
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Dandan Guo
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Chengyuan Song
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Jun Ma
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Yiming Liu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
7
|
Cheng X, Wu T, Han L, Sun T, Huang G. Association between added sugars intake and Parkinson's disease status in U.S. adults: a cross-sectional study from NHANES 1990-2020. Arch Public Health 2024; 82:225. [PMID: 39593073 PMCID: PMC11590255 DOI: 10.1186/s13690-024-01445-8] [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: 07/17/2024] [Accepted: 11/09/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Added sugars intake is common among individuals with Parkinson's disease (PD), yet the link between added sugars intake and PD is not well understood. Our study aims to investigate the association between added sugars intake and PD. METHODS This study utilized data from the National Health and Nutrition Examination Survey (NHANES) from1990 to 2020. Added sugars intake was estimated based on a 24-hour dietary recall from participants. Multivariable logistic regression analysis was employed to explore the relationship between added sugars intake and the prevalence of PD. Restricted cubic spline (RCS) was used to explore the nonlinear association between added sugars intake and PD. To further observe whether the conclusions were consistent across different subgroups, we conducted subgroup analyses to investigate the association of added sugars intake with PD in different populations. RESULTS The study included 12,489 participants, of which 100 had PD. When weighted, the data represented 136,959,144 participants. The study revealed a positive association between added sugars intake and the prevalence of PD. In multivariable regression models adjusted for all confounding factors, compared with the lowest quartile of added sugars intake, the third quartile (OR = 2.99; 95% CI: 1.43-6.26) and those consuming more than 25% of their calories from added sugars (OR = 3.34; 95% CI: 1.03-10.86) had the highest risk of PD. The RCS curve showed an L-shaped nonlinear association between added sugars intake and PD. Two-segment linear regression by sex revealed that PD prevalence in women was linearly related to sugar intake (nonlinear P = 0.465), while men exhibited an L-shaped nonlinear relationship (nonlinear P = 0.03). Additionally, subgroup analysis showed that alcohol consumption and diabetes significantly influenced the association between added sugars intake and the prevalence of PD. CONCLUSION These results highlight a positive association between added sugars intake and the prevalence of PD, particularly among women, heavy drinkers, and individuals with diabetes.
Collapse
Affiliation(s)
- Xuehua Cheng
- Department of Traditional Chinese Medicine (TCM) Geriatrics, Huadong Hospital, Fudan University, Shanghai, 200040, China
| | - Tao Wu
- Department of Traditional Chinese Medicine (TCM) Geriatrics, Huadong Hospital, Fudan University, Shanghai, 200040, China
| | - Li Han
- Department of Traditional Chinese Medicine (TCM) Geriatrics, Huadong Hospital, Fudan University, Shanghai, 200040, China
| | - Tong Sun
- Department of Neonatology, Jiuting Hospital, Shanghai, 201615, China.
| | - Guoxin Huang
- Department of Evidence-Based Medicine Center, People's Hospital, Hubei University of Medicine, Xiangyang No.1, Xiangyang, 441000, China.
| |
Collapse
|
8
|
Pei J, Zhang C, Zhang Q, Yu H, Yuan H, Guo Y, Shen H, Liu H, Wang C, Meng F, Yu C, Tie J, Chen X, Wu X, Zhang G, Wang X. Probiotics alleviate chronic ethanol exposure-induced anxiety-like behavior and hippocampal neuroinflammation in male mice through gut microbiota-derived extracellular vesicles. J Nanobiotechnology 2024; 22:730. [PMID: 39578835 PMCID: PMC11585232 DOI: 10.1186/s12951-024-03017-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Probiotics can colonize both the human and animal bodies and consist of active microorganisms that are beneficial to health. The use of probiotics has been shown to alleviate certain neurological diseases and disturbances in gut microbiota resulting from chronic ethanol exposure. Research indicates that probiotics can influence the nervous system via the microbial-gut-brain axis, wherein extracellular vesicles secreted by the gut microbiota play a significant role in this process. RESULTS In this study, we first established a 30-day ethanol exposure and probiotic gavage mouse model, both of which influenced behavior and the composition of gut microbiota. We then extracted gut microbiota-derived extracellular vesicles from the feces of these model mice and injected them into new mice via the tail vein to assess the role of each set of extracellular vesicles. The results indicated that the extracellular vesicles derived from the intestinal microbiota in the ethanol group induced anxiety-like behavior and hippocampal neuroinflammation in the recipient mice. In contrast, the extracellular vesicles secreted by the gut microbiota from the probiotic group mitigated the anxiety-like behavior and neuroinflammation induced by ethanol-influenced extracellular vesicles. CONCLUSIONS Our study demonstrates that extracellular vesicles secreted by the gut microbiota can influence the nervous system via the microbial-gut-brain axis. Furthermore, we found that the extracellular vesicles secreted by the gut microbiota from the probiotic group exert a beneficial therapeutic effect on anxiety and hippocampal neuroinflammation.
Collapse
Affiliation(s)
- Jiaxin Pei
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, Liaoning, People's Republic of China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang, Liaoning, People's Republic of China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, People's Republic of China
| | - Chaoxu Zhang
- Department of Hematology, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Qian Zhang
- Department of Health Statistics, School of Public Health, China Medical University, Shenyang, Liaoning, People's Republic of China
- Department of Reproductive Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning, People's Republic of China
| | - Hao Yu
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, Liaoning, People's Republic of China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang, Liaoning, People's Republic of China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, People's Republic of China
| | - Huiya Yuan
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang, Liaoning, People's Republic of China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, People's Republic of China
- Department of Forensic Analytical Toxicology, China Medical University School of Forensic Medicine, Shenyang, 110122, Liaoning, People's Republic of China
| | - Yufu Guo
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, Liaoning, People's Republic of China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang, Liaoning, People's Republic of China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, People's Republic of China
| | - Hui Shen
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, Liaoning, People's Republic of China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang, Liaoning, People's Republic of China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, People's Republic of China
| | - Hao Liu
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, Liaoning, People's Republic of China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang, Liaoning, People's Republic of China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, People's Republic of China
| | - Changliang Wang
- The People's Procuratorate of Liaoning Province Judicial Authentication Center, Shenyang, Liaoning, People's Republic of China
- Collaborative Laboratory of Intelligentized Forensic Science (CLIFS), Shenyang, Liaoning, People's Republic of China
| | - Fanyue Meng
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, Liaoning, People's Republic of China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang, Liaoning, People's Republic of China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, People's Republic of China
- Department of Morphology, Medical College of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Chenyang Yu
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, Liaoning, People's Republic of China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang, Liaoning, People's Republic of China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, People's Republic of China
| | - Jinming Tie
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, Liaoning, People's Republic of China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang, Liaoning, People's Republic of China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, People's Republic of China
| | - Xiaohuan Chen
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, Liaoning, People's Republic of China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang, Liaoning, People's Republic of China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, People's Republic of China
| | - Xu Wu
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, Liaoning, People's Republic of China.
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang, Liaoning, People's Republic of China.
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, People's Republic of China.
| | - Guohua Zhang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, Liaoning, People's Republic of China.
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang, Liaoning, People's Republic of China.
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, People's Republic of China.
| | - Xiaolong Wang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, Liaoning, People's Republic of China.
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang, Liaoning, People's Republic of China.
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
9
|
Gonzalez HC, Misare KR, Mendenhall TT, Wolf BJ, Mulholland PJ, Gordon KL, Hartman JH. Transgenic expression of human cytochrome P450 2E1 in C. elegans and rat PC-12 cells sensitizes to ethanol-induced locomotor and mitochondrial effects. Biochem Biophys Res Commun 2024; 734:150735. [PMID: 39357336 PMCID: PMC11540131 DOI: 10.1016/j.bbrc.2024.150735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/17/2024] [Accepted: 09/21/2024] [Indexed: 10/04/2024]
Abstract
Chronic alcohol (ethanol) use is increasing in the United States and has been linked to numerous health issues in multiple organ systems including neurological dysfunction and diseases. Ethanol toxicity is mainly driven by the metabolite acetaldehyde, which is generated through three pathways: alcohol dehydrogenase (ADH2), catalase (CAT), and cytochrome P450 2E1 (CYP2E1). ADH2, while the main ethanol clearance pathway in the liver, is not expressed in the mammalian brain, resulting in CAT and CYP2E1 driving local metabolism of ethanol in the central nervous system. CYP2E1 is known to generate reactive metabolites and reactive oxygen species and localizes to the mitochondria (mtCYP2E1) and endoplasmic reticulum (erCYP2E1). We sought to understand the consequences of mtCYP2E1 and erCYP2E1 in the nervous system during acute ethanol exposure. To answer this question, we generated transgenic Caenorhabditis elegans roundworms expressing human CYP2E1 in the mitochondria, endoplasmic reticulum, or both and exposed them to ethanol. We found that at lower concentrations, wild-type and mtCYP2E1-expressing worms had a small but significant inhibition of locomotion, whereas the erCYP2E1-expressing worms showed protection from this inhibition. At higher doses, all strains had reduced locomotion, but the erCYP2E1-expressing worms recovered faster than wild-type controls. CYP2E1 expression, regardless of organellar targeting, reduced mitochondrial respiration in response to ethanol. Similarly, transgenic expression of CYP2E1 in either organelle in PC-12 rat neuronal cell lines sensitized them to ethanol-induced cell death. Together, these findings suggest that subcellular localization of CYP2E1 impacts behavioral effects of ethanol and should be further studied in the mammalian central nervous system.
Collapse
Affiliation(s)
- Hyland C Gonzalez
- Dept of Biochemistry and Molecular Biology, Medical University of South Carolina, 173 Ashley Ave. BSB 501 | MSC 509, Charleston, SC, 29425, USA
| | - Kelly R Misare
- Dept of Biochemistry and Molecular Biology, Medical University of South Carolina, 173 Ashley Ave. BSB 501 | MSC 509, Charleston, SC, 29425, USA
| | - Tsultrim T Mendenhall
- Dept of Biochemistry and Molecular Biology, Medical University of South Carolina, 173 Ashley Ave. BSB 501 | MSC 509, Charleston, SC, 29425, USA
| | - Bethany J Wolf
- Dept of Public Health Sciences, Medical University of South Carolina, 135 Cannon St., Charleston, SC, 29425, USA
| | - Patrick J Mulholland
- Dept of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - Kacy L Gordon
- Dept of Biology, The University of North Carolina at Chapel Hill, 316 Fordham Hall, Chapel Hill, NC, 27599, USA; UNC Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, 450 West Dr, Chapel Hill, NC, 27599, USA
| | - Jessica H Hartman
- Dept of Biochemistry and Molecular Biology, Medical University of South Carolina, 173 Ashley Ave. BSB 501 | MSC 509, Charleston, SC, 29425, USA; Dept of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Ave. BSB 501 | MSC 509, Charleston, SC, 29425, USA.
| |
Collapse
|
10
|
Bouajila N, Domenighetti C, Aubin HJ, Naassila M. Alcohol consumption and its association with cancer, cardiovascular, liver and brain diseases: a systematic review of Mendelian randomization studies. FRONTIERS IN EPIDEMIOLOGY 2024; 4:1385064. [PMID: 39574800 PMCID: PMC11578756 DOI: 10.3389/fepid.2024.1385064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 10/14/2024] [Indexed: 11/24/2024]
Abstract
Background The health effects of alcohol consumption, particularly regarding potential protective benefits of light to moderate intake compared to abstinence, remain a subject of ongoing debate. However, epidemiological studies face limitations due to imprecise exposure measurements and the potential for bias through residual confounding and reverse causation. To address these limitations, we conducted a systematic review of Mendelian Randomization (MR) studies examining the causal relationship between alcohol consumption and cancers, cardiovascular, liver, and neurological diseases. Methodology We searched PubMed, ScienceDirect and Embase and Europe PMC up to 05/2024 for MR studies investigating the association of genetically predicted alcohol consumption with cancers, cardiovascular, liver and neurological diseases. We assessed methodological quality based on key elements of the MR design a genetic association studies tool. Results We included 70 MR studies that matched our inclusion criteria. Our review showed a significant association of alcohol consumption with multiple cancers such as oral and oropharyngeal, esophageal, colorectal cancers, hepatocellular carcinoma and cutaneous melanoma. While the available studies did not consistently confirm the adverse or protective effects of alcohol on other cancers, such as lung cancer, as suggested by observational studies. Additionally, MR studies confirmed a likely causal effect of alcohol on the risk of hypertension, atrial fibrillation, myocardial infraction and vessels disease. However, there was no evidence to support the protective effects of light to moderate alcohol consumption on cognitive function, Alzheimer's disease, and amyotrophic lateral sclerosis, as reported in observational studies while our review revealed an increased risk of epilepsy and multiple sclerosis. The available studies provided limited results on the link between alcohol consumption and liver disease. Conclusions Despite the valuable insights into the causal relationship between alcohol consumption and various health outcomes that MR studies provided, it is worth noting that the inconsistent ability of genetic instrumental variables to distinguish between abstainers, light and moderate drinkers makes it difficult to differentiate between U or J-shaped vs. linear relationships between exposure and outcome. Additional research is necessary to establish formal quality assessment tools for MR studies and to conduct more studies in diverse populations, including non-European ancestries. Systematic Review Registration www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021246154, Identifier: PROSPERO (CRD42021246154).
Collapse
Affiliation(s)
- Naouras Bouajila
- Inserm Unit UMRS 1247, University of Picardie Jules Verne, Amiens, France
| | - Cloé Domenighetti
- UVSQ, Univ. Paris-Sud, Inserm, Team “Exposome, Heredity, Cancer, and Health”, CESP, University of Paris-Saclay, Villejuif, France
| | - Henri-Jean Aubin
- Department of Psychiatry and Addictology, Paul-Brousse Hospital, AP-HP, Center for Epidemiology and Population Health Research (CESP), Inserm 1018, University of Paris-Saclay, Villejuif, France
| | - Mickael Naassila
- Inserm Unit UMRS 1247, University of Picardie Jules Verne, Amiens, France
| |
Collapse
|
11
|
Mercer A, Sancandi M, Maclatchy A, Lange S. Brain-Region-Specific Differences in Protein Citrullination/Deimination in a Pre-Motor Parkinson's Disease Rat Model. Int J Mol Sci 2024; 25:11168. [PMID: 39456949 PMCID: PMC11509057 DOI: 10.3390/ijms252011168] [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: 09/16/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
The detection of early molecular mechanisms and potential biomarkers in Parkinson's disease (PD) remains a challenge. Recent research has pointed to novel roles for post-translational citrullination/deimination caused by peptidylarginine deiminases (PADs), a family of calcium-activated enzymes, in the early stages of the disease. The current study assessed brain-region-specific citrullinated protein targets and their associated protein-protein interaction networks alongside PAD isozymes in the 6-hydroxydopamine (6-OHDA) induced rat model of pre-motor PD. Six brain regions (cortex, hippocampus, striatum, midbrain, cerebellum and olfactory bulb) were compared between controls/shams and the pre-motor PD model. For all brain regions, there was a significant difference in citrullinated protein IDs between the PD model and the controls. Citrullinated protein hits were most abundant in cortex and hippocampus, followed by cerebellum, midbrain, olfactory bulb and striatum. Citrullinome-associated pathway enrichment analysis showed correspondingly considerable differences between the six brain regions; some were overlapping for controls and PD, some were identified for the PD model only, and some were identified in control brains only. The KEGG (Kyoto Encyclopedia of Genes and Genomes) pathways identified in PD brains only were associated with neurological, metabolic, immune and hormonal functions and included the following: "Axon guidance"; "Spinocerebellar ataxia"; "Hippo signalling pathway"; "NOD-like receptor signalling pathway"; "Phosphatidylinositol signalling system"; "Rap1 signalling pathway"; "Platelet activation"; "Yersinia infection"; "Fc gamma R-mediated phagocytosis"; "Human cytomegalovirus infection"; "Inositol phosphate metabolism"; "Thyroid hormone signalling pathway"; "Progesterone-mediated oocyte maturation"; "Oocyte meiosis"; and "Choline metabolism in cancer". Some brain-region-specific differences were furthermore observed for the five PAD isozymes (PADs 1, 2, 3, 4 and 6), with most changes in PAD 2, 3 and 4 when comparing control and PD brain regions. Our findings indicate that PAD-mediated protein citrullination plays roles in metabolic, immune, cell signalling and neurodegenerative disease-related pathways across brain regions in early pre-motor stages of PD, highlighting PADs as targets for future therapeutic avenues.
Collapse
Affiliation(s)
- Audrey Mercer
- Department of Pharmacology, UCL School of Pharmacy, London WC1N 1AX, UK; (A.M.); (M.S.)
| | - Marco Sancandi
- Department of Pharmacology, UCL School of Pharmacy, London WC1N 1AX, UK; (A.M.); (M.S.)
| | - Amy Maclatchy
- Pathobiology and Extracellular Vesicles Research Group, School of Life Sciences, University of Westminster, London W1W 6XH, UK;
| | - Sigrun Lange
- Pathobiology and Extracellular Vesicles Research Group, School of Life Sciences, University of Westminster, London W1W 6XH, UK;
| |
Collapse
|
12
|
Kang S, Lee J, Ali DN, Choi S, Nesbitt J, Min PH, Trushina E, Choi DS. Low to moderate ethanol exposure reduces astrocyte-induced neuroinflammatory signaling and cognitive decline in presymptomatic APP/PS1 mice. Sci Rep 2024; 14:23989. [PMID: 39402264 PMCID: PMC11473946 DOI: 10.1038/s41598-024-75202-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
Alcohol use disorder has been associated with the development of neurodegenerative diseases, including Alzheimer's disease (AD). However, recent studies demonstrate that moderate alcohol consumption may be protective against dementia and cognitive decline. We examined astrocyte function, low-density lipoprotein (LDL) receptor-related protein 1 (LRP1), and the NF-κB p65 and IKK-α/β signaling pathways in modulating neuroinflammation and amyloid beta (Aβ) deposition. We assessed apolipoprotein E (ApoE) in the brain of APP/PS1 mice using IHC and ELISA in response to low to moderate ethanol exposure (MEE). First, to confirm the intracerebral distribution of ApoE, we co-stained with GFAP, a marker for astrocytes that biosynthesize ApoE. We sought to investigate whether the ethanol-induced upregulation of LRP1 could potentially inhibit the activity of IL-1β and TNF-α induced IKK-α/β towards NF-κB p65, resulting in a reduction of pro-inflammatory cytokines. To evaluate the actual Aβ load in the brains of APP/PS1 mice, we performed with a specific antibody Aβ (Thioflavin S) on both air- and ethanol-exposed groups, subsequently analyzing Aβ levels. We also measured glucose uptake using 18F- fluorodeoxyglucose (FDG)-positron emission tomography (PET). Finally, we investigated whether MEE induced cognitive and memory changes using the Y maze, noble object recognition test, and Morris water maze. Our findings demonstrate that MEE reduced astrocytic glial fibrillary acidic protein (GFAP) and ApoE levels in the cortex and hippocampus in presymptomatic APP/PS1 mice. Interestingly, increased LRP1 protein expression was accompanied by dampening the IKK-α/β-NF-κB p65 pathway, resulting in decreased IL-1β and TNF-α levels in male mice. Notably, female mice show reduced levels of anti-inflammatory cytokines IL-4, and IL-10 without altering IL-1β and TNF-α concentrations. In both males and females, Aβ plaques, a hallmark of AD, were reduced in the cortex and hippocampus of APP/PS1 mice exposed to ethanol starting at pre-symptomatic stage. Consistently, MEE increased FDG-PET-based brain activities and normalized cognitive and memory deficits in the APP/PS1 mice. Our findings suggest that MEE may benefit AD pathology via modulating LRP1 expression, potentially reducing neuroinflammation and attenuating Aβ deposition. Our study implies that reduced astrocyte-derived ApoE and LDL cholesterol levels are critical for attenuating AD pathology.
Collapse
Affiliation(s)
- Shinwoo Kang
- Department of Molecular Pharmacology and Experimental Therapeutics, Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Pharmacology College of Medicine, Soonchunhyang University, 22 Soonchunhyango-ro, Ansan, Chungcheongnam-do, 31508, South Korea
| | - Jeyeon Lee
- Department of Radiology, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN, 55905, USA
| | - Dina N Ali
- Department of Molecular Pharmacology and Experimental Therapeutics, Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA
| | - Sun Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jarred Nesbitt
- Department of Neurology, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN, 55905, USA
| | - Paul H Min
- Department of Radiology, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN, 55905, USA
| | - Eugenia Trushina
- Department of Molecular Pharmacology and Experimental Therapeutics, Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Neurology, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN, 55905, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA.
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN, 55905, USA.
- Neuroscience Program, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
13
|
Huang L, Liu M, Tang J, Gong Z, Li Z, Yang Y, Zhang M. The role of ALDH2 rs671 polymorphism and C-reactive protein in the phenotypes of male ALS patients. Front Neurosci 2024; 18:1397991. [PMID: 39290715 PMCID: PMC11405379 DOI: 10.3389/fnins.2024.1397991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/07/2024] [Indexed: 09/19/2024] Open
Abstract
Background The aldehyde dehydrogenase 2 (ALDH2) rs671 (A) allele has been implicated in neurodegeneration, potentially through oxidative and inflammatory pathways. The study aims to investigate the effects of the ALDH2 rs671 (A) allele and high sensitivity C-reactive protein (hs-CRP) on the clinical phenotypes of amyotrophic lateral sclerosis (ALS) in male and female patients. Methods Clinical data and ALDH2 rs671 genotype of 143 ALS patients, including 85 males and 58 females, were collected from January 2018 to December 2022. All patients underwent assessment using the Chinese version of the Edinburgh Cognitive and Behavioral ALS Screen (ECAS). Complete blood count and metabolic profiles were measured. Clinical and laboratory parameters were compared between carriers and non-carriers of the rs671 (A) allele in males and females, respectively. The significant parameters and rs671 (A) Allele were included in multivariate linear regression models to identify potential contributors to motor and cognitive impairment. Mediation analysis was employed to evaluate any mediation effects. Results Male patients carrying rs671 (A) allele exhibited higher levels of hs-CRP than non-carriers (1.70 mg/L vs. 0.50 mg/L, p = 0.006). The rs671 (A) allele was identified as an independent risk factor for faster disease progression only in male patients (β = 0.274, 95% CI = 0.048-0.499, p = 0.018). The effect of the rs671 (A) allele on the executive function in male patients was fully mediated by hs-CRP (Indirect effect = -1.790, 95% CI = -4.555--0.225). No effects of the rs671 (A) allele or hs-CRP were observed in female ALS patients. The effects of the ALDH2 rs671 (A) allele and the mediating role of hs-CRP in male patients remained significant in the sensitivity analyses. Conclusion The ALDH2 rs671 (A) allele contributed to faster disease progression and hs-CRP mediated cognitive impairment in male ALS patients.
Collapse
Affiliation(s)
- Lifang Huang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mao Liu
- Department of Neurology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States
| | - Jiahui Tang
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Zhenxiang Gong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zehui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Yang
- Department of Neurology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Min Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Neurology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
14
|
Karatayev O, Collier AD, Targoff SR, Leibowitz SF. Neurological Disorders Induced by Drug Use: Effects of Adolescent and Embryonic Drug Exposure on Behavioral Neurodevelopment. Int J Mol Sci 2024; 25:8341. [PMID: 39125913 PMCID: PMC11313660 DOI: 10.3390/ijms25158341] [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: 06/18/2024] [Revised: 07/10/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
Clinical studies demonstrate that the risk of developing neurological disorders is increased by overconsumption of the commonly used drugs, alcohol, nicotine and cannabis. These drug-induced neurological disorders, which include substance use disorder (SUD) and its co-occurring emotional conditions such as anxiety and depression, are observed not only in adults but also with drug use during adolescence and after prenatal exposure to these drugs, and they are accompanied by long-lasting disturbances in brain development. This report provides overviews of clinical and preclinical studies, which confirm these adverse effects in adolescents and the offspring prenatally exposed to the drugs and include a more in-depth description of specific neuronal systems, their neurocircuitry and molecular mechanisms, affected by drug exposure and of specific techniques used to determine if these effects in the brain are causally related to the behavioral disturbances. With analysis of further studies, this review then addresses four specific questions that are important for fully understanding the impact that drug use in young individuals can have on future pregnancies and their offspring. Evidence demonstrates that the adverse effects on their brain and behavior can occur: (1) at low doses with short periods of drug exposure during pregnancy; (2) after pre-conception drug use by both females and males; (3) in subsequent generations following the initial drug exposure; and (4) in a sex-dependent manner, with drug use producing a greater risk in females than males of developing SUDs with emotional conditions and female offspring after prenatal drug exposure responding more adversely than male offspring. With the recent rise in drug use by adolescents and pregnant women that has occurred in association with the legalization of cannabis and increased availability of vaping tools, these conclusions from the clinical and preclinical literature are particularly alarming and underscore the urgent need to educate young women and men about the possible harmful effects of early drug use and to seek novel therapeutic strategies that might help to limit drug use in young individuals.
Collapse
Affiliation(s)
| | | | | | - Sarah F. Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, USA; (O.K.); (S.R.T.)
| |
Collapse
|
15
|
Gomes TM, Sousa P, Campos C, Perestrelo R, Câmara JS. Secondary Bioactive Metabolites from Foods of Plant Origin as Theravention Agents against Neurodegenerative Disorders. Foods 2024; 13:2289. [PMID: 39063373 PMCID: PMC11275480 DOI: 10.3390/foods13142289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/14/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Neurodegenerative disorders (NDDs) such as Alzheimer's (AD) and Parkinson's (PD) are on the rise, robbing people of their memories and independence. While risk factors such as age and genetics play an important role, exciting studies suggest that a diet rich in foods from plant origin may offer a line of defense. These kinds of foods, namely fruits and vegetables, are packed with a plethora of powerful bioactive secondary metabolites (SBMs), including terpenoids, polyphenols, glucosinolates, phytosterols and capsaicinoids, which exhibit a wide range of biological activities including antioxidant, antidiabetic, antihypertensive, anti-Alzheimer's, antiproliferative, and antimicrobial properties, associated with preventive effects in the development of chronic diseases mediated by oxidative stress such as type 2 diabetes mellitus, respiratory diseases, cancer, cardiovascular diseases, and NDDs. This review explores the potential of SBMs as theravention agents (metabolites with therapeutic and preventive action) against NDDs. By understanding the science behind plant-based prevention, we may be able to develop new strategies to promote brain health and prevent the rise in NDDs. The proposed review stands out by emphasizing the integration of multiple SBMs in plant-based foods and their potential in preventing NDDs. Previous research has often focused on individual compounds or specific foods, but this review aims to present a comprehensive fingerprint of how a diet rich in various SBMs can synergistically contribute to brain health. The risk factors related to NDD development and the diagnostic process, in addition to some examples of food-related products and medicinal plants that significantly reduce the inhibition of acetylcholinesterase (AChE), butyrylcholinesterase (BChE), and β-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1), are highlighted.
Collapse
Affiliation(s)
- Telma Marisa Gomes
- CQM—Centro de Química da Madeira, NPRG, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal; (T.M.G.); (P.S.); (C.C.); (R.P.)
| | - Patrícia Sousa
- CQM—Centro de Química da Madeira, NPRG, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal; (T.M.G.); (P.S.); (C.C.); (R.P.)
| | - Catarina Campos
- CQM—Centro de Química da Madeira, NPRG, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal; (T.M.G.); (P.S.); (C.C.); (R.P.)
| | - Rosa Perestrelo
- CQM—Centro de Química da Madeira, NPRG, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal; (T.M.G.); (P.S.); (C.C.); (R.P.)
| | - José S. Câmara
- CQM—Centro de Química da Madeira, NPRG, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal; (T.M.G.); (P.S.); (C.C.); (R.P.)
- Departamento de Química, Faculdade de Ciências Exatas e Engenharia, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal
| |
Collapse
|
16
|
Kang J, Deng YT, Wu BS, Liu WS, Li ZY, Xiang S, Yang L, You J, Gong X, Jia T, Yu JT, Cheng W, Feng J. Whole exome sequencing analysis identifies genes for alcohol consumption. Nat Commun 2024; 15:5777. [PMID: 38982111 PMCID: PMC11233704 DOI: 10.1038/s41467-024-50132-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 06/26/2024] [Indexed: 07/11/2024] Open
Abstract
Alcohol consumption is a heritable behavior seriously endangers human health. However, genetic studies on alcohol consumption primarily focuses on common variants, while insights from rare coding variants are lacking. Here we leverage whole exome sequencing data across 304,119 white British individuals from UK Biobank to identify protein-coding variants associated with alcohol consumption. Twenty-five variants are associated with alcohol consumption through single variant analysis and thirteen genes through gene-based analysis, ten of which have not been reported previously. Notably, the two unreported alcohol consumption-related genes GIGYF1 and ANKRD12 show enrichment in brain function-related pathways including glial cell differentiation and are strongly expressed in the cerebellum. Phenome-wide association analyses reveal that alcohol consumption-related genes are associated with brain white matter integrity and risk of digestive and neuropsychiatric diseases. In summary, this study enhances the comprehension of the genetic architecture of alcohol consumption and implies biological mechanisms underlying alcohol-related adverse outcomes.
Collapse
Affiliation(s)
- Jujiao Kang
- Institute of Science and Technology for Brain-Inspired Intelligence (ISTBI), Fudan University, Shanghai, 200433, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, 200433, China
| | - Yue-Ting Deng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200433, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200433, China
| | - Wei-Shi Liu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200433, China
| | - Ze-Yu Li
- Institute of Science and Technology for Brain-Inspired Intelligence (ISTBI), Fudan University, Shanghai, 200433, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, 200433, China
| | - Shitong Xiang
- Institute of Science and Technology for Brain-Inspired Intelligence (ISTBI), Fudan University, Shanghai, 200433, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, 200433, China
| | - Liu Yang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200433, China
| | - Jia You
- Institute of Science and Technology for Brain-Inspired Intelligence (ISTBI), Fudan University, Shanghai, 200433, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, 200433, China
| | - Xiaohong Gong
- School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Tianye Jia
- Institute of Science and Technology for Brain-Inspired Intelligence (ISTBI), Fudan University, Shanghai, 200433, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, 200433, China
- Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- School of Psychology, University of Southampton, Southampton, UK
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200433, China.
| | - Wei Cheng
- Institute of Science and Technology for Brain-Inspired Intelligence (ISTBI), Fudan University, Shanghai, 200433, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, 200433, China.
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200433, China.
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Zhejiang, China.
| | - Jianfeng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence (ISTBI), Fudan University, Shanghai, 200433, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, 200433, China.
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Zhejiang, China.
- Department of Computer Science, University of Warwick, Coventry, CV4 7AL, UK.
| |
Collapse
|
17
|
Chen H, Wang X, Chang Z, Zhang J, Xie D. Evidence for genetic causality between iron homeostasis and Parkinson's disease: A two-sample Mendelian randomization study. J Trace Elem Med Biol 2024; 84:127430. [PMID: 38484633 DOI: 10.1016/j.jtemb.2024.127430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/07/2024] [Accepted: 03/09/2024] [Indexed: 05/27/2024]
Abstract
BACKGROUND Parkinson's disease (PD) is a degenerative disease of the central nervous system, and its specific etiology is still unclear. At present, it is believed that the main pathological basis is the reduction of dopamine concentration in the brain striatum. Although many previous studies have believed that iron as an important nutrient element participates in the occurrence and development of PD, whether there is a causal correlation between total iron binding capacity(TIBC), transferring saturation(TSAT), ferritin and serum iron in iron homeostasis indicators and PD, there has been a lack of effective genetic evidence. METHODS We used Mendelian randomization (MR) as an analytical method to effectively evaluate the genetic association between exposure and outcome, based on the largest genome-wide association study (GWAS) data to date. By using randomly assigned genetic instrumental variables (SNPs, Single Nucleotide Polymorphisms) that are not affected by any causal relationship, we effectively evaluated the causal relationship between iron homeostasis indicators and PD while controlling for confounding factors. RESULTS By coordinated analysis of 86 SNPs associated with iron homeostasis markers and 12,858,066 SNPs associated with PD, a total of 56 SNPs were finally screened for genome-wide significance of iron homeostasis associated with PD. The results of inverse variance weighting(IVW) analysis suggested that iron( β = - 0.524; 95%cl=-0.046 to -0.002; P=0.032) was considered to have a genetic causal relationship with PD. Cochran's Q, Egger intercept and MR-PRESSO global tests did not detect the existence of heterogeneity and pleiotropy (P>0.05). Mr Steiger directionality test further confirmed our estimation of the potential causal direction of iron and PD (P=0.001). In addition, TIBC (β=-0.142; 95%Cl=-0.197-0.481; P=0.414), TSAT (β=-0.316; 95%Cl=-0.861-0.229; P=0.255), and ferritin (β=-0.387; 95%Cl=-1.179-0.405; P=0.338) did not have genetic causal relationships with PD, and the results were not heterogeneous and pleiotropic (P>0.05). In addition, TIBC (β=-0.142; 95%Cl=-0.197-0.481; P=0.414), TSAT (β=-0.316; 95%Cl=-0.861-0.229; P=0.255), and ferritin (β=-0.101; 95%Cl=--0.987 to -0.405; P=0.823) did not have genetic causal relationships with PD, and the results were not heterogeneous and pleiotropic (P>0.05). TIBC (P=0.008), TSAT (P=0.000) and ferritin (P=0.013) were all consistent with the estimation of MR Steiger directivity test. CONCLUSION Our study found that among the four iron homeostasis markers, there was a genetic causal association between serum iron and PD, and the serum iron level was negatively correlated with the risk of PD. In addition, TIBC, TSAT, ferritin had no genetic causal relationship with PD.
Collapse
Affiliation(s)
- Hong Chen
- Anhui University of Chinese Medicine, Hefei 230038, China
| | - Xie Wang
- Anhui University of Chinese Medicine, Hefei 230038, China
| | - Ze Chang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100089, China
| | - Juan Zhang
- Department of Neurology, the First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei 230031, China.
| | - Daojun Xie
- Department of Neurology, the First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei 230031, China
| |
Collapse
|
18
|
Heidari N, Hajikarim-Hamedani A, Heidari A, Ghane Y, Ashabi G, Zarrindast MR, Sadat-Shirazi MS. Alcohol: Epigenome alteration and inter/transgenerational effect. Alcohol 2024; 117:27-41. [PMID: 38508286 DOI: 10.1016/j.alcohol.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024]
Abstract
While DNA serves as the fundamental genetic blueprint for an organism, it is not a static entity. Gene expression, the process by which genetic information is utilized to create functional products like proteins, can be modulated by a diverse range of environmental factors. Epigenetic mechanisms, including DNA methylation, histone modification, and microRNAs, play a pivotal role in mediating the intricate interplay between the environment and gene expression. Intriguingly, alterations in the epigenome have the potential to be inherited across generations. Alcohol use disorder (AUD) poses significant health issues worldwide. Alcohol has the capability to induce changes in the epigenome, which can be inherited by offspring, thus impacting them even in the absence of direct alcohol exposure. This review delves into the impact of alcohol on the epigenome, examining how its effects vary based on factors such as the age of exposure (adolescence or adulthood), the duration of exposure (chronic or acute), and the specific sample collected (brain, blood, or sperm). The literature underscores that alcohol exposure can elicit diverse effects on the epigenome during different life stages. Furthermore, compelling evidence from human and animal studies demonstrates that alcohol induces alterations in epigenome content, affecting both the brain and blood. Notably, rodent studies suggest that these epigenetic changes can result in lasting phenotype alterations that extend across at least two generations. In conclusion, the comprehensive literature analysis supports the notion that alcohol exposure induces lasting epigenetic alterations, influencing the behavior and health of future generations. This knowledge emphasizes the significance of addressing the potential transgenerational effects of alcohol and highlights the importance of preventive measures to minimize the adverse impact on offspring.
Collapse
Affiliation(s)
- Nazila Heidari
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Amirhossein Heidari
- Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Yekta Ghane
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghorbangol Ashabi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Zarrindast
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
19
|
Singh N, Nandy SK, Jyoti A, Saxena J, Sharma A, Siddiqui AJ, Sharma L. Protein Kinase C (PKC) in Neurological Health: Implications for Alzheimer's Disease and Chronic Alcohol Consumption. Brain Sci 2024; 14:554. [PMID: 38928554 PMCID: PMC11201589 DOI: 10.3390/brainsci14060554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Protein kinase C (PKC) is a diverse enzyme family crucial for cell signalling in various organs. Its dysregulation is linked to numerous diseases, including cancer, cardiovascular disorders, and neurological problems. In the brain, PKC plays pivotal roles in synaptic plasticity, learning, memory, and neuronal survival. Specifically, PKC's involvement in Alzheimer's Disease (AD) pathogenesis is of significant interest. The dysregulation of PKC signalling has been linked to neurological disorders, including AD. This review elucidates PKC's pivotal role in neurological health, particularly its implications in AD pathogenesis and chronic alcohol addiction. AD, characterised by neurodegeneration, implicates PKC dysregulation in synaptic dysfunction and cognitive decline. Conversely, chronic alcohol consumption elicits neural adaptations intertwined with PKC signalling, exacerbating addictive behaviours. By unravelling PKC's involvement in these afflictions, potential therapeutic avenues emerge, offering promise for ameliorating their debilitating effects. This review navigates the complex interplay between PKC, AD pathology, and alcohol addiction, illuminating pathways for future neurotherapeutic interventions.
Collapse
Affiliation(s)
- Nishtha Singh
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology, and Management Sciences, Solan 173229, Himachal Pradesh, India; (N.S.); (A.S.)
| | - Shouvik Kumar Nandy
- School of Pharmacy, Techno India University, Sector-V, Kolkata 700091, West Bengal, India;
| | - Anupam Jyoti
- Department of Life Science, Parul Institute of Applied Science, Parul University, Vadodara 391760, Gujarat, India;
| | - Juhi Saxena
- Department of Biotechnology, Parul Institute of Technology, Parul University, Vadodara 391760, Gujarat, India;
| | - Aditi Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology, and Management Sciences, Solan 173229, Himachal Pradesh, India; (N.S.); (A.S.)
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Hail, Hail 55476, Saudi Arabia
| | - Lalit Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology, and Management Sciences, Solan 173229, Himachal Pradesh, India; (N.S.); (A.S.)
| |
Collapse
|
20
|
Wang Z, Yang X, Li H, Wang S, Liu Z, Wang Y, Zhang X, Chen Y, Xu Q, Xu J, Wang Z, Wang J. Bidirectional two-sample Mendelian randomization analyses support causal relationships between structural and diffusion imaging-derived phenotypes and the risk of major neurodegenerative diseases. Transl Psychiatry 2024; 14:215. [PMID: 38806463 PMCID: PMC11133432 DOI: 10.1038/s41398-024-02939-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 05/10/2024] [Accepted: 05/16/2024] [Indexed: 05/30/2024] Open
Abstract
Previous observational investigations suggest that structural and diffusion imaging-derived phenotypes (IDPs) are associated with major neurodegenerative diseases; however, whether these associations are causal remains largely uncertain. Herein we conducted bidirectional two-sample Mendelian randomization analyses to infer the causal relationships between structural and diffusion IDPs and major neurodegenerative diseases using common genetic variants-single nucleotide polymorphism (SNPs) as instrumental variables. Summary statistics of genome-wide association study (GWAS) for structural and diffusion IDPs were obtained from 33,224 individuals in the UK Biobank cohort. Summary statistics of GWAS for seven major neurodegenerative diseases were obtained from the largest GWAS for each disease to date. The forward MR analyses identified significant or suggestively statistical causal effects of genetically predicted three structural IDPs on Alzheimer's disease (AD), frontotemporal dementia (FTD), and multiple sclerosis. For example, the reduction in the surface area of the left superior temporal gyrus was associated with a higher risk of AD. The reverse MR analyses identified significantly or suggestively statistical causal effects of genetically predicted AD, Lewy body dementia (LBD), and FTD on nine structural and diffusion IDPs. For example, LBD was associated with increased mean diffusivity in the right superior longitudinal fasciculus and AD was associated with decreased gray matter volume in the right ventral striatum. Our findings might contribute to shedding light on the prediction and therapeutic intervention for the major neurodegenerative diseases at the neuroimaging level.
Collapse
Affiliation(s)
- Zirui Wang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xuan Yang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Department of Radiology, Jining No.1 People's Hospital, Jining, Shandong, 272000, China
| | - Haonan Li
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Siqi Wang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Zhixuan Liu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yaoyi Wang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xingyu Zhang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yayuan Chen
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Qiang Xu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jiayuan Xu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Zengguang Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Junping Wang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
21
|
Alam MJ, Rahman MH, Hossain MA, Hoque MR, Aktaruzzaman M. Bioinformatics and Systems Biology Approaches to Identify the Synergistic Effects of Alcohol Use Disorder on the Progression of Neurological Diseases. Neuroscience 2024; 543:65-82. [PMID: 38401711 DOI: 10.1016/j.neuroscience.2024.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 02/26/2024]
Abstract
Clinical investigations showed that individuals with Alcohol Use Disorder (AUD) have worse Neurological Disease (ND) development, pointing to possible pathogenic relationships between AUD and NDs. It remains difficult to identify risk factors that are predisposing between AUD and NDs. In order to fix these issues, we created the bioinformatics pipeline and network-based approaches for employing unbiased methods to discover genes abnormally stated in both AUD and NDs and to pinpoint some of the common molecular pathways that might underlie AUD and ND interaction. We found 100 differentially expressed genes (DEGs) in both the AUD and ND patient's tissue samples. The most important Gene Ontology (GO) terms and metabolic pathways, including positive control of cytotoxicity caused by T cells, proinflammatory responses, antigen processing and presentation, and platelet-triggered interactions with vascular and circulating cell pathways were then extracted using the overlapped DEGs. Protein-protein interaction analysis was used to identify hub proteins, including CCL2, IL1B, TH, MYCN, HLA-DRB1, SLC17A7, and HNF4A, in the pathways that have been reported as playing a function in these disorders. We determined several TFs (HNF4A, C4A, HLA-B, SNCA, HLA-DMB, SLC17A7, HLA-DRB1, HLA-C, HLA-A, and HLA-DPB1) and potential miRNAs (hsa-mir-34a-5p, hsa-mir-34c-5p, hsa-mir-449a, hsa-mir-155-5p, and hsa-mir-1-3p) were crucial for regulating the expression of AUD and ND which could serve as prospective targets for treatment. Our methodologies discovered unique putative biomarkers that point to the interaction between AUD and various neurological disorders, as well as pathways that could one day be the focus of therapeutic intervention.
Collapse
Affiliation(s)
- Md Jahangir Alam
- Department of Computer Science and Engineering, Islamic University, Kushtia 7003, Bangladesh; Center for Advanced Bioinformatics and Artificial Intelligence Research, Islamic University, Kushtia 7003, Bangladesh
| | - Md Habibur Rahman
- Department of Computer Science and Engineering, Islamic University, Kushtia 7003, Bangladesh; Center for Advanced Bioinformatics and Artificial Intelligence Research, Islamic University, Kushtia 7003, Bangladesh.
| | - Md Arju Hossain
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail 1902, Bangladesh; Department of Microbiology, Primeasia University, Banani, Dhaka 1213, Bangladesh
| | - Md Robiul Hoque
- Department of Computer Science and Engineering, Islamic University, Kushtia 7003, Bangladesh
| | - Md Aktaruzzaman
- Department of Computer Science and Engineering, Islamic University, Kushtia 7003, Bangladesh
| |
Collapse
|
22
|
Smith GC, Griffith KR, Sicher AR, Brockway DF, Proctor EA, Crowley NA. MODERATE ALCOHOL CONSUMPTION INDUCES LASTING IMPACTS ON PREFRONTAL CORTICAL SIGNALING IN MICE. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587955. [PMID: 38617243 PMCID: PMC11014573 DOI: 10.1101/2024.04.03.587955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Both alcohol use disorder (AUD) and Alzheimer's Disease and Related Dementias (ADRD) appear to include disruption in the balance of excitation and inhibition in the cortex, but their potential interactions are unclear. We examined the effect of moderate voluntary binge alcohol consumption on the aged, pre-disease neuronal environment by measuring intrinsic excitability and spontaneous neurotransmission on prefrontal cortical pyramidal (excitatory, glutamatergic) and non-pyramidal (inhibitory, GABAergic) neurons following a prolonged period of abstinence from alcohol in mice. Results highlight that binge alcohol consumption has lasting impacts on the electrophysiological properties of prefrontal cortical neurons. A profound increase in excitatory events onto layer 2/3 non-pyramidal neurons following alcohol consumption was seen, along with altered intrinsic excitability of pyramidal neurons, which could have a range of effects on Alzheimer's Disease progression in humans. These results indicate that moderate voluntary alcohol influences the pre-disease environment in aging and highlight the need for further mechanistic investigation into this risk factor.
Collapse
Affiliation(s)
- Grace C Smith
- Department of Biology, The Pennsylvania State University, University Park, PA, USA 16802
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA 16802
| | - Keith R Griffith
- Department of Biology, The Pennsylvania State University, University Park, PA, USA 16802
| | - Avery R Sicher
- Department of Biology, The Pennsylvania State University, University Park, PA, USA 16802
- Neuroscience Graduate Program, Huck Institute of the Life Sciences, The Pennsylvania State University, University Park, PA, USA 16802
| | - Dakota F Brockway
- Department of Biology, The Pennsylvania State University, University Park, PA, USA 16802
- Neuroscience Graduate Program, Huck Institute of the Life Sciences, The Pennsylvania State University, University Park, PA, USA 16802
| | - Elizabeth A Proctor
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA 16802
- Departments of Neurosurgery Penn State College of Medicine, Hershey PA, USA 17033; and Engineering Science and Mechanics, University Park, PA, USA 16802
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Nicole A Crowley
- Department of Biology, The Pennsylvania State University, University Park, PA, USA 16802
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA 16802
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|
23
|
Wang X, Wang J, Chen Y, Qian X, Luo S, Wang X, Ma C, Ge W. The aldehyde dehydrogenase 2 rs671 variant enhances amyloid β pathology. Nat Commun 2024; 15:2594. [PMID: 38519490 PMCID: PMC10959958 DOI: 10.1038/s41467-024-46899-0] [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: 08/11/2023] [Accepted: 03/12/2024] [Indexed: 03/25/2024] Open
Abstract
In the ALDH2 rs671 variant, a guanine changes to an adenine, resulting in a dramatic decrease in the catalytic activity of the enzyme. Population-based data are contradictory about whether this variant increases the risk of Alzheimer's disease. In East Asian populations, the prevalence of the ALDH2 rs671 variant is 30-50%, making the National Human Brain Bank for Development and Function (the largest brain bank in East Asia) an important resource to explore the link between the ALDH2 rs671 polymorphism and Alzheimer's disease pathology. Here, using 469 postmortem brains, we find that while the ALDH2 rs671 variant is associated with increased plaque deposits and a higher Aβ40/42 ratio, it is not an independent risk factor for Alzheimer's disease. Mechanistically, we show that lower ALDH2 activity leads to 4-HNE accumulation in the brain. The (R)-4-HNE enantiomer adducts to residue Lys53 of C99, favoring Aβ40 generation in the Golgi apparatus. Decreased ALDH2 activity also lowers inflammatory factor secretion, as well as amyloid β phagocytosis and spread in brains of patients with Alzheimer's disease. We thus define the relationship between the ALDH2 rs671 polymorphism and amyloid β pathology, and find that ALDH2 rs671 is a key regulator of Aβ40 or Aβ42 generation.
Collapse
Affiliation(s)
- Xia Wang
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Jiayu Wang
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Yashuang Chen
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Xiaojing Qian
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, National Human Brain Bank for Development and Function, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Shiqi Luo
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Xue Wang
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, National Human Brain Bank for Development and Function, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Chao Ma
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, National Human Brain Bank for Development and Function, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
| | - Wei Ge
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
| |
Collapse
|
24
|
Seemiller LR, Flores-Cuadra J, Griffith KR, Smith GC, Crowley NA. Alcohol and stress exposure across the lifespan are key risk factors for Alzheimer's Disease and cognitive decline. Neurobiol Stress 2024; 29:100605. [PMID: 38268931 PMCID: PMC10806346 DOI: 10.1016/j.ynstr.2024.100605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/11/2023] [Accepted: 01/03/2024] [Indexed: 01/26/2024] Open
Abstract
Alzheimer's Disease and related dementias (ADRD) are an increasing threat to global health initiatives. Efforts to prevent the development of ADRD require understanding behaviors that increase and decrease risk of neurodegeneration and cognitive decline, in addition to uncovering the underlying biological mechanisms behind these effects. Stress exposure and alcohol consumption have both been associated with increased risk for ADRD in human populations. However, our ability to understand causal mechanisms of ADRD requires substantial preclinical research. In this review, we summarize existing human and animal research investigating the connections between lifetime stress and alcohol exposures and ADRD.
Collapse
Affiliation(s)
- Laurel R. Seemiller
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Julio Flores-Cuadra
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
- Neuroscience Graduate Program, Huck Institute of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Keith R. Griffith
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Grace C. Smith
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Nicole A. Crowley
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- Neuroscience Graduate Program, Huck Institute of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
25
|
Walsh M, Uretsky M, Tripodis Y, Nowinski CJ, Rasch A, Bruce H, Ryder M, Martin BM, Palmisano JN, Katz DI, Dwyer B, Daneshvar DH, Walley AY, Kim TW, Goldstein LE, Stern RA, Alvarez VE, Huber BR, McKee AC, Stein TD, Mez J, Alosco ML. Clinical and Neuropathological Correlates of Substance Use in American Football Players. J Alzheimers Dis 2024; 101:971-986. [PMID: 39269838 DOI: 10.3233/jad-240300] [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: 09/15/2024]
Abstract
Background Chronic traumatic encephalopathy (CTE) is a neurodegenerative tauopathy more frequently found in deceased former football players. CTE has heterogeneous clinical presentations with multifactorial causes. Previous literature has shown substance use (alcohol/drug) can contribute to Alzheimer's disease and related tauopathies pathologically and clinically. Objective To examine the association between substance use and clinical and neuropathological endpoints of CTE. Methods Our sample included 429 deceased male football players. CTE was neuropathologically diagnosed. Informant interviews assessed features of substance use and history of treatment for substance use to define indicators: history of substance use treatment (yes vs no, primary variable), alcohol severity, and drug severity. Outcomes included scales that were completed by informants to assess cognition (Cognitive Difficulties Scale, BRIEF-A Metacognition Index), mood (Geriatric Depression Scale-15), behavioral regulation (BRIEF-A Behavioral Regulation Index, Barratt Impulsiveness Scale-11), functional ability (Functional Activities Questionnaire), as well as CTE status and cumulative p-tau burden. Regression models tested associations between substance use indicators and outcomes. Results Of the 429 football players (mean age = 62.07), 313 (73%) had autopsy confirmed CTE and 100 (23%) had substance use treatment history. Substance use treatment and alcohol/drug severity were associated with measures of behavioral regulation (FDR-p-values<0.05, ΔR2 = 0.04-0.18) and depression (FDR-p-values<0.05, ΔR2 = 0.02-0.05). Substance use indicators had minimal associations with cognitive scales, whereas p-tau burden was associated with all cognitive scales (p-values <0.05). Substance use treatment had no associations with neuropathological endpoints (FDR-p-values>0.05). Conclusions Among deceased football players, substance use was common and associated with clinical symptoms.
Collapse
Affiliation(s)
- Michael Walsh
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Madeline Uretsky
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Yorghos Tripodis
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Christopher J Nowinski
- Department of Neurology, Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Concussion Legacy Foundation, Boston, MA, USA
| | - Abigail Rasch
- Department of Neurology, Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Hannah Bruce
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Megan Ryder
- Department of Neurology, Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Brett M Martin
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA, USA
| | - Joseph N Palmisano
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA, USA
| | - Douglas I Katz
- Department of Neurology, Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Braintree Rehabilitation Hospital, Braintree, MA, USA
| | - Brigid Dwyer
- Department of Neurology, Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Braintree Rehabilitation Hospital, Braintree, MA, USA
| | - Daniel H Daneshvar
- Department of Neurology, Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Massachusetts General Hospital, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Boston, MA, USA
| | - Alexander Y Walley
- Grayken Center for Addiction, Clinical Addiction Research and Education Unit, Section of General Internal Medicine, Boston Medical Center, and Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Theresa W Kim
- Grayken Center for Addiction, Clinical Addiction Research and Education Unit, Section of General Internal Medicine, Boston Medical Center, and Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Lee E Goldstein
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Radiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Departments of Biomedical, Electrical & Computer Engineering, Boston University College of Engineering, Boston, MA, USA
| | - Robert A Stern
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurosurgery, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Victor E Alvarez
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- U.S. Department of Veteran Affairs, VA Boston Healthcare System, Jamaica Plain, MA, USA
- National Center for PTSD, VA Boston Healthcare, Jamaica Plain, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Bertrand Russell Huber
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- U.S. Department of Veteran Affairs, VA Boston Healthcare System, Jamaica Plain, MA, USA
- National Center for PTSD, VA Boston Healthcare, Jamaica Plain, MA, USA
| | - Ann C McKee
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- U.S. Department of Veteran Affairs, VA Boston Healthcare System, Jamaica Plain, MA, USA
- National Center for PTSD, VA Boston Healthcare, Jamaica Plain, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Thor D Stein
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- U.S. Department of Veteran Affairs, VA Boston Healthcare System, Jamaica Plain, MA, USA
- National Center for PTSD, VA Boston Healthcare, Jamaica Plain, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jesse Mez
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Michael L Alosco
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
26
|
Ahmed T, Zhang P, Kumar K. Equilibrium of Dietary Patterns Between Alzheimer's Disease Patients and Healthy People: A Comprehensive Analysis Using Multiple Factor Analysis and Classification Modeling. J Alzheimers Dis 2024; 97:777-790. [PMID: 38189748 DOI: 10.3233/jad-230634] [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: 01/09/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is a particular type of dementia that currently lacks a definitive treatment and cure. It is possible to reduce the risk of developing AD and mitigate its severity through modifications to one's lifestyle, regular diet, and alcohol-drinking habits. OBJECTIVE The objective of this study is to examine the daily dietary patterns of individuals with AD compared to healthy controls, with a focus on nutritional balance and its impact on AD. METHODS This study incorporated multiple-factor analysis (MFA) to evaluate dietary patterns and employed Random Forest (RF) classifier and Sparse Logistic Regression (SLR) for Variable Importance analysis to identify food items significantly associated with AD. RESULTS MFA revealed trends in the data and a strong correlation (Lg = 0.92, RV = 0.65) between the daily consumption of processed food and meat items in AD patients. In contrast, no significant relationship was found for any daily consumed food categories within the healthy control (HC) group. Food items such as meat pie, hamburger, ham, sausages, beef, capsicum, and cabbage were identified as important variables associated with AD in RF and SLR analyses. CONCLUSIONS The findings from MFA indicated that the diversity or equilibrium of daily diet might play a potential role in AD development. RF and SLR classifications exhibit among the processed foods, especially deli meats and food made with meat items, are associated with AD.
Collapse
Affiliation(s)
- Tahera Ahmed
- Centre for Data Analytics, Bond Business School, Bond University, Gold Coast, Australia
| | - Ping Zhang
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Kuldeep Kumar
- Centre for Data Analytics, Bond Business School, Bond University, Gold Coast, Australia
| |
Collapse
|
27
|
Rodríguez-Ayala M, Donat-Vargas C, Moreno-Franco B, Mérida DM, Ramón Banegas J, Rodríguez-Artalejo F, Guallar-Castillón P. Association of a healthy beverage score with total mortality in the adult population of Spain: A nationwide cohort study. PLoS Med 2024; 21:e1004337. [PMID: 38261590 PMCID: PMC10805278 DOI: 10.1371/journal.pmed.1004337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/21/2023] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Despite the substantial evidence of the relationship between diet and mortality, the role of beverage consumption patterns is not well known. The aim of this study was to assess the association of the adherence to a Healthy Beverage Score (HBS) and all-cause mortality in a representative sample of the Spanish adult population. METHODS AND FINDINGS We conducted an observational cohort study using data from the Study on Nutrition and Cardiovascular Risk in Spain (ENRICA), which included 12,161 community-dwelling individuals aged ≥18 years recruited in 2008 to 2010 and followed until January 2022. At baseline, food consumption was collected using a validated diet history. The HBS consists of 7 items, each of which is scored from 1 to 4 (highest adherence). The HBS ranges from 7 to 28 points with a higher score representing a healthier pattern. Adherence was assigned as a higher consumption of low-fat milk, and coffee and tea, a lower consumption of whole-fat milk, no consumption of fruit juice, artificially sweetened beverages, or sugar-sweetened beverages, and no or moderate consumption of alcohol. Total mortality was ascertained by linkage to the Spanish National Death Index. Statistical analyses were performed with Cox models and adjusted for the main confounders, including sociodemographic, lifestyle, dietary variables, and morbidity. After a mean follow-up of 12.5 years (SD: 1.7; range: 0.5 to 12.9), a total of 967 deaths occurred. For all-cause mortality, the fully adjusted hazard ratio (HR) for the highest versus lowest sex-specific quartiles of HBS was 0.72 (95% confidence interval [0.57, 0.91], p linear-trend = 0.015), corresponding to an 8.3% reduction in the absolute risk of death. A linear relationship between the risk of death and the adherence to the HBS was observed using restricted cubic splines. The results were robust to sensitivity analyses. The main limitation was that repeated measurements on beverage consumption were not available and beverage consumption could have changed during follow-up. CONCLUSIONS In this study, we observed that higher adherence to the HBS was associated with lower total mortality. Adherence to a healthy beverage pattern could play a role in the prevention of premature mortality.
Collapse
Affiliation(s)
- Montserrat Rodríguez-Ayala
- Department of Preventive Medicine and Public Health, School of Medicine, Universidad Autónoma de Madrid and CIBERESP (CIBER of Epidemiology and Public Health), Madrid, Spain
- Department of Microbiology and Parasitology, Hospital Universitario La Paz, Madrid, Spain
| | - Carolina Donat-Vargas
- Department of Preventive Medicine and Public Health, School of Medicine, Universidad Autónoma de Madrid and CIBERESP (CIBER of Epidemiology and Public Health), Madrid, Spain
- ISGlobal, Campus Mar., Barcelona, Spain
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Belén Moreno-Franco
- Instituto de Investigación Sanitaria (IIS) Aragón, Hospital Universitario Miguel Servet, Zaragoza, Spain
- CIBERCV (CIBER of Cardiovascular), Instituto de Salud Carlos III, Madrid, Spain
| | - Diana María Mérida
- Department of Preventive Medicine and Public Health, School of Medicine, Universidad Autónoma de Madrid and CIBERESP (CIBER of Epidemiology and Public Health), Madrid, Spain
| | - José Ramón Banegas
- Department of Preventive Medicine and Public Health, School of Medicine, Universidad Autónoma de Madrid and CIBERESP (CIBER of Epidemiology and Public Health), Madrid, Spain
| | - Fernando Rodríguez-Artalejo
- Department of Preventive Medicine and Public Health, School of Medicine, Universidad Autónoma de Madrid and CIBERESP (CIBER of Epidemiology and Public Health), Madrid, Spain
- IMDEA-Food Institute, CEI UAM+CSIC., Madrid, Spain
| | - Pilar Guallar-Castillón
- Department of Preventive Medicine and Public Health, School of Medicine, Universidad Autónoma de Madrid and CIBERESP (CIBER of Epidemiology and Public Health), Madrid, Spain
- IMDEA-Food Institute, CEI UAM+CSIC., Madrid, Spain
| |
Collapse
|
28
|
Kang S, Lee J, Choi S, Nesbitt J, Min PH, Trushina E, Choi DS. Moderate ethanol exposure reduces astrocyte-induced neuroinflammatorysignaling and cognitive decline in presymptomatic APP/PS1 mice. RESEARCH SQUARE 2023:rs.3.rs-3627637. [PMID: 38077051 PMCID: PMC10705690 DOI: 10.21203/rs.3.rs-3627637/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Background Alcohol use disorder (AUD) has been associated with the development of neurodegenerative diseases, including Alzheimer's disease (AD). However, recent studies demonstrate that moderate alcohol consumption may be protective against dementia and cognitive decline. Methods We examined astrocyte function, low-density lipoprotein (LDL) receptor-related protein 1 (LRP1), and the NF-κB p65 and IKK-α/β signaling pathways in modulating neuroinflammation and amyloid beta (Aβ) deposition. We assessed apolipoprotein E (ApoE) in the mouse brain using IHC and ELISA in response to moderate ethanol exposure (MEE). First, to confirm the intracerebral distribution of ApoE, we co-stained with GFAP, a marker for astrocytes that biosynthesize ApoE. We sought to investigate whether the ethanol-induced upregulation of LRP1 could potentially inhibit the activity of IL-1β and TNF-α induced IKK-α/β towards NF-κB p65, resulting in a reduction of pro-inflammatory cytokines. To evaluate the actual Aβ load in the brains of APP/PS1 mice, we performed with a specific antibody Aβ (Thioflavin S) on both air- and ethanol-exposed groups, subsequently analyzing Aβ levels. We also measured glucose uptake activity using 18F-FDG in APP/PS1 mice. Finally, we investigated whether MEE induced cognitive and memory changes using the Y maze, noble objective recognition (NOR) test, and Morris water maze (MWM). Results Our findings demonstrate that MEE reduced astrocytic glial fibrillary acidic protein (GFAP) and ApoE levels in the cortex and hippocampus in presymptomatic APP/PS1 mice. Interestingly, increased LRP1 protein expression is accompanied by dampening the IKK-α/β-NF-κB p65 pathway, resulting in decreased IL-1β and TNF-α levels in male mice. Notably, female mice show reduced anti-inflammatory cytokines, IL-4, and IL-10 levels without altering IL-1β and TNF-α concentrations. In both males and females, Aβ plaques, a hallmark of AD, were reduced in the cortex and hippocampus of ethanol-exposed presymptomatic APP/PS1 mice. Consistently, MEE increased fluorodeoxyglucose (FDG)-positron emission tomography (PET)-based brain activities and normalized cognitive and memory deficits in the APP/PS1 mice. Conclusions Our findings suggest that MEE may benefit AD pathology via modulating LRP1 expression, potentially reducing neuroinflammation and attenuating Aβ deposition. Our study implies that reduced astrocyte derived ApoE and LDL cholesterol levels are critical for attenuating AD pathology.
Collapse
Affiliation(s)
| | - Jeyeon Lee
- Mayo Clinic College of Medicine, and Science
| | - Sun Choi
- Mayo Clinic College of Medicine, and Science
| | | | - Paul H Min
- Mayo Clinic College of Medicine, and Science
| | | | | |
Collapse
|
29
|
Al-Ayari EA, Shehata MG, El-Hadidi M, Shaalan MG. In silico SNP prediction of selected protein orthologues in insect models for Alzheimer's, Parkinson's, and Huntington's diseases. Sci Rep 2023; 13:18986. [PMID: 37923901 PMCID: PMC10624829 DOI: 10.1038/s41598-023-46250-5] [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: 08/22/2023] [Accepted: 10/30/2023] [Indexed: 11/06/2023] Open
Abstract
Alzheimer's, Parkinson's, and Huntington's are the most common neurodegenerative diseases that are incurable and affect the elderly population. Discovery of effective treatments for these diseases is often difficult, expensive, and serendipitous. Previous comparative studies on different model organisms have revealed that most animals share similar cellular and molecular characteristics. The meta-SNP tool includes four different integrated tools (SIFT, PANTHER, SNAP, and PhD-SNP) was used to identify non synonymous single nucleotide polymorphism (nsSNPs). Prediction of nsSNPs was conducted on three representative proteins for Alzheimer's, Parkinson's, and Huntington's diseases; APPl in Drosophila melanogaster, LRRK1 in Aedes aegypti, and VCPl in Tribolium castaneum. With the possibility of using insect models to investigate neurodegenerative diseases. We conclude from the protein comparative analysis between different insect models and nsSNP analyses that D. melanogaster is the best model for Alzheimer's representing five nsSNPs of the 21 suggested mutations in the APPl protein. Aedes aegypti is the best model for Parkinson's representing three nsSNPs in the LRRK1 protein. Tribolium castaneum is the best model for Huntington's disease representing 13 SNPs of 37 suggested mutations in the VCPl protein. This study aimed to improve human neural health by identifying the best insect to model Alzheimer's, Parkinson's, and Huntington's.
Collapse
Affiliation(s)
- Eshraka A Al-Ayari
- Entomology Department, Faculty of Science, Ain Shams University, Cairo, Egypt.
| | - Magdi G Shehata
- Entomology Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Mohamed El-Hadidi
- Bioinformatics Group, Center for Informatics Sciences (CIS), School of Information Technology and Computer Science (ITCS) , Nile University, Giza, Egypt
| | - Mona G Shaalan
- Entomology Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
30
|
Khan MAS, Chang SL. Alcohol and the Brain-Gut Axis: The Involvement of Microglia and Enteric Glia in the Process of Neuro-Enteric Inflammation. Cells 2023; 12:2475. [PMID: 37887319 PMCID: PMC10605902 DOI: 10.3390/cells12202475] [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: 07/16/2023] [Revised: 09/25/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023] Open
Abstract
Binge or chronic alcohol consumption causes neuroinflammation and leads to alcohol use disorder (AUD). AUD not only affects the central nervous system (CNS) but also leads to pathologies in the peripheral and enteric nervous systems (ENS). Thus, understanding the mechanism of the immune signaling to target the effector molecules in the signaling pathway is necessary to alleviate AUD. Growing evidence shows that excessive alcohol consumption can activate neuroimmune cells, including microglia, and change the status of neurotransmitters, affecting the neuroimmune system. Microglia, like peripheral macrophages, are an integral part of the immune defense and represent the reticuloendothelial system in the CNS. Microglia constantly survey the CNS to scavenge the neuronal debris. These cells also protect parenchymal cells in the brain and spinal cord by repairing nerve circuits to keep the nervous system healthy against infectious and stress-derived agents. In an activated state, they become highly dynamic and mobile and can modulate the levels of neurotransmitters in the CNS. In several ways, microglia, enteric glial cells, and macrophages are similar in terms of causing inflammation. Microglia also express most of the receptors that are constitutively present in macrophages. Several receptors on microglia respond to the inflammatory signals that arise from danger-associated molecular patterns (DAMPs), pathogen-associated molecular patterns (PAMPs), endotoxins (e.g., lipopolysaccharides), and stress-causing molecules (e.g., alcohol). Therefore, this review article presents the latest findings, describing the roles of microglia and enteric glial cells in the brain and gut, respectively, and their association with neurotransmitters, neurotrophic factors, and receptors under the influence of binge and chronic alcohol use, and AUD.
Collapse
Affiliation(s)
- Mohammed A. S. Khan
- Department of Neurosurgery, Brigham Hospital for Children, Harvard Medical School, Boston, MA 02115, USA;
| | - Sulie L. Chang
- Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, NJ 07079, USA
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07079, USA
| |
Collapse
|
31
|
Ferreira LK, Filgueiras Meireles JF, de Oliveira Gomes GA, Caputo Ferreira ME. Development and Psychometric Evaluation of a Lifestyle Evaluation Instrument for Older Adults. Percept Mot Skills 2023; 130:1901-1923. [PMID: 37286477 DOI: 10.1177/00315125231182173] [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: 06/09/2023]
Abstract
Brazil is experiencing an accelerated period of population aging, with important implications for individuals, families, and the society at large. Lifestyles for the elderly can have both positive and negative health effects and are characterized by behaviors that individuals constantly reproduce in day-to-day life. However, few assessment instruments are aimed at evaluating lifestyles, and this has hampered research development. Thus, our aim in this study was to develop and evaluate the psychometric qualities of a new instrument to assess older adults' lifestyles. We developed this single scale to assess the lifestyles of older men and women using the sequential mixed method. Individuals over 60 years old of both sexes participated in this study of several phases. In Phase 1, we produced a 96-item single scale instrument, based on a literature review, previous scales, and qualitative research. In Phase 2, we verified the scale's content validity with help from 12 experts and 20 members of a target audience, aged between 60-84 years, deleting some items and reworking others. In Phase 3, we analyzed the psychometric qualities of the new scale in a sample of 700 older adults, aged 60 and above, from the five regions of Brazil, using both exploratory and confirmatory factor analysis. Our completed "Older Adult Lifestyle Scale" (OALS) is composed of 19 items divided into four subscales. The OALS has shown good psychometric qualities for Brazilian older adults over 60 years of age, and we can now recommend its use in this population.
Collapse
|
32
|
Jung SY, Chun S, Cho EB, Han K, Yoo J, Yeo Y, Yoo JE, Jeong SM, Min JH, Shin DW. Changes in smoking, alcohol consumption, and the risk of Parkinson's disease. Front Aging Neurosci 2023; 15:1223310. [PMID: 37771519 PMCID: PMC10525683 DOI: 10.3389/fnagi.2023.1223310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/21/2023] [Indexed: 09/30/2023] Open
Abstract
Objective There have been no studies on the association between changes in smoking and alcohol consumption or combined changes in smoking and alcohol consumption frequencies and PD risk. To assess the influence of changes in smoking and alcohol consumption on the risk of Parkinson's disease (PD). Methods National Health Insurance Service (NHIS) database between January 2009 to December 2011 was analyzed. A total of 3,931,741 patients were included. Study participants were followed up for the incidence of PD until December 2017. Results Compared to the sustained non-smokers, sustained light smokers (adjusted hazard ratio [aHR] 0.80, 95% confidence interval [CI] 0.75-0.85), sustained moderate smokers (aHR 0.54, 95% CI 0.47-0.61), and sustained heavy smokers (aHR 0.49, 95% CI 0.44-0.55) had a lower risk of PD. Compared to those who sustained non-drinking, sustained light drinkers (aHR 0.85 95% CI 0.89-0.91), sustained moderate drinkers (aHR 0.68, 95% CI 0.60-0.78), and sustained heavy drinkers (aHR 0.77, 95% CI 0.68-0.87) showed decreased risk of PD. Among non-drinkers, those who started drinking to a light level were at decreased risk of PD (aHR 0.84, 95% CI 0.77-0.91). Among non-smoking and non-drinking participants, those who initiated smoking only (aHR 0.78, 95% CI 0.70-0.86), drinking only (aHR 0.77, 95% CI 0.68-0.87), and both smoking and drinking (aHR 0.69, 95% CI 0.58-0.82) showed decreased risk of PD. Conclusion Smoking is associated with decreased risk of PD with a dose-response relationship. Alcohol consumption at a light level may also be associated with decreased risk of PD. Further studies are warranted to find the possible mechanisms for the protective effects of smoking and drinking on PD, which may present insights into the etiology of PD.
Collapse
Affiliation(s)
- Se Young Jung
- Department of Family Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Digital Healthcare, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Sohyun Chun
- International Healthcare Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Eun Bin Cho
- Department of Neurology, College of Medicine, Gyeongsang Institute of Health Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Juhwan Yoo
- Department of Biostatistics, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yohwan Yeo
- Department of Family Medicine/Supportive Care Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jung Eun Yoo
- Department of Family Medicine, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Su Min Jeong
- Department of Family Medicine/Supportive Care Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ju-Hong Min
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Dong Wook Shin
- Department of Family Medicine/Supportive Care Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Clinical Study Design and Evaluation, Samsung Advanced Institute of Health Science and Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
- Center for Wireless and Population Health Systems, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
33
|
Chavan RS, Supalkar KV, Sadar SS, Vyawahare NS. Animal models of Alzheimer's disease: An originof innovativetreatments and insight to the disease's etiology. Brain Res 2023; 1814:148449. [PMID: 37302570 DOI: 10.1016/j.brainres.2023.148449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/04/2023] [Accepted: 06/05/2023] [Indexed: 06/13/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder. The main pathogenic features are the development and depositionof senile plaques and neurofibrillary tangles in brain. Recent developments in the knowledge of the pathophysiological mechanisms behind Alzheimer's disease and other cognitive disorders have suggested new approaches to treatment development. These advancements have been significantly aided by the use of animal models, which are also essential for the assessment of therapies. Various approaches as transgenic animal model, chemical models, brain injury are used. This review will presentAD pathophysiology and emphasize several Alzheimer like dementia causingchemical substances, transgenic animal model and stereotaxy in order to enhance our existing knowledge of their mechanism of AD induction, dose, and treatment duration.
Collapse
Affiliation(s)
- Ritu S Chavan
- D. Y. Patil College of Pharmacy, Akurdi, Pune 411044, Maharashtra, India.
| | - Krishna V Supalkar
- D. Y. Patil College of Pharmacy, Akurdi, Pune 411044, Maharashtra, India
| | - Smeeta S Sadar
- D. Y. Patil College of Pharmacy, Akurdi, Pune 411044, Maharashtra, India
| | - Niraj S Vyawahare
- D. Y. Patil College of Pharmacy, Akurdi, Pune 411044, Maharashtra, India
| |
Collapse
|
34
|
Seike T, Chen CH, Mochly-Rosen D. Impact of common ALDH2 inactivating mutation and alcohol consumption on Alzheimer's disease. Front Aging Neurosci 2023; 15:1223977. [PMID: 37693648 PMCID: PMC10483235 DOI: 10.3389/fnagi.2023.1223977] [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: 05/16/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Aldehyde dehydrogenase 2 (ALDH2) is an enzyme found in the mitochondrial matrix that plays a central role in alcohol and aldehyde metabolism. A common ALDH2 polymorphism in East Asians descent (called ALDH2*2 or E504K missense variant, SNP ID: rs671), present in approximately 8% of the world's population, has been associated with a variety of diseases. Recent meta-analyses support the relationship between this ALDH2 polymorphism and Alzheimer's disease (AD). And AD-like pathology observed in ALDH2-/- null mice and ALDH2*2 overexpressing transgenic mice indicate that ALDH2 deficiency plays an important role in the pathogenesis of AD. Recently, the worldwide increase in alcohol consumption has drawn attention to the relationship between heavy alcohol consumption and AD. Of potential clinical significance, chronic administration of alcohol in ALDH2*2/*2 knock-in mice exacerbates the pathogenesis of AD-like symptoms. Therefore, ALDH2 polymorphism and alcohol consumption likely play an important role in the onset and progression of AD. Here, we review the data on the relationship between ALDH2 polymorphism, alcohol, and AD, and summarize what is currently known about the role of the common ALDH2 inactivating mutation, ALDH2*2, and alcohol in the onset and progression of AD.
Collapse
Affiliation(s)
| | | | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
35
|
Huang LY, Ou YN, Yang YX, Wang ZT, Tan L, Yu JT. Associations of cardiovascular risk factors and lifestyle behaviors with neurodegenerative disease: a Mendelian randomization study. Transl Psychiatry 2023; 13:267. [PMID: 37488110 PMCID: PMC10366095 DOI: 10.1038/s41398-023-02553-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/26/2023] Open
Abstract
Previous observational studies reported that midlife clustering of cardiovascular risk factors and lifestyle behaviors were associated with neurodegenerative disease; however, these findings might be biased by confounding and reverse causality. This study aimed to investigate the causal associations of cardiovascular risk factors and lifestyle behaviors with neurodegenerative disease, using the two-sample Mendelian randomization design. Genetic variants for the modifiable risk factors and neurodegenerative disease were extracted from large-scale genome-wide association studies. The inverse-variance weighted method was used as the main analysis method, and MR-Egger regression and leave-one-out analyses were performed to identify potential violations. Genetically predicted diastolic blood pressure (DBP: OR per 1 mmHg, 0.990 [0.979-1.000]), body mass index (BMI: OR per 1 SD, 0.880 [0.825-0.939]), and educational level (OR per 1 SD, 0.698 [0.602-0.810]) were associated with lower risk of late-onset Alzheimer's disease (LOAD), while genetically predicted low-density lipoprotein (LDL: OR per 1 SD, 1.302 [1.066-1.590]) might increase LOAD risk. Genetically predicted exposures (including LDL and BMI) applied to familial AD showed the same effect. The association of LDL was also found with Amyotrophic lateral sclerosis (ALS) (LDL: OR per 1 SD, 1.180 [1.080-1.289]). This MR analysis showed that LDL, BMI, BP, and educational level were causally related to AD; a significant association between LDL and ALS risk, as well as the potential effect of sleep duration on PD risk, were also revealed. Targeting these modifiable factors was a promising strategy of neurodegenerative disease prevention.
Collapse
Affiliation(s)
- Liang-Yu Huang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yu-Xiang Yang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
36
|
Vejux A. Cell Death and Neurodegenerative Diseases: Mechanisms and Cytoprotective Molecules. Int J Mol Sci 2023; 24:11465. [PMID: 37511220 PMCID: PMC10380922 DOI: 10.3390/ijms241411465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/11/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
A neurodegenerative disease is a pathological condition affecting neurons, condemning them to death [...].
Collapse
Affiliation(s)
- Anne Vejux
- Team Bio-PeroxIL, "Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism" (EA7270), Université de Bourgogne Franche-Comté, UFR Sciences Vie Terre et Environnement, 21000 Dijon, France
| |
Collapse
|
37
|
Zhang C, Chen H, Rodriguez Y, Ma X, Swerdlow RH, Zhang J, Ding WX. A perspective on autophagy and transcription factor EB in Alcohol-Associated Alzheimer's disease. Biochem Pharmacol 2023; 213:115576. [PMID: 37127251 PMCID: PMC11009931 DOI: 10.1016/j.bcp.2023.115576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/03/2023]
Abstract
Alzheimer's disease (AD) is the most common form of progressive dementia and there is no truly efficacious treatment. Accumulating evidence indicates that impaired autophagic function for removal of damaged mitochondria and protein aggregates such as amyloid and tau protein aggregates may contribute to the pathogenesis of AD. Epidemiologic studies have implicated alcohol abuse in promoting AD, yet the underlying mechanisms are poorly understood. In this review, we discuss mechanisms of selective autophagy for mitochondria and protein aggregates and how these mechanisms are impaired by aging and alcohol consumption. We also discuss potential genetic and pharmacological approaches for targeting autophagy/mitophagy, as well as lysosomal and mitochondrial biogenesis, for the potential prevention and treatment of AD.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Hao Chen
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Yssa Rodriguez
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Xiaowen Ma
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Russell H Swerdlow
- Department of Neurology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jianhua Zhang
- Department of Pathology, Division of Molecular Cellular Pathology, University of Alabama at Birmingham, 901 19th street South, Birmingham, AL 35294, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; Department of Internal Medicine, Division of Gastroenterology, Hepatology & Motility, The University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
38
|
Jiang X, Yan C, Zhang H, Chen L, Jiang R, Zheng K, Jin W, Ma H, Liu X, Dong M. Oral Probiotic Expressing Human Ethanol Dehydrogenase Attenuates Damage Caused by Acute Alcohol Consumption in Mice. Microbiol Spectr 2023; 11:e0429422. [PMID: 37039510 PMCID: PMC10269551 DOI: 10.1128/spectrum.04294-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/24/2023] [Indexed: 04/12/2023] Open
Abstract
Alcohol is an essential drug in human life with multiple medical functions, but excessive alcohol intake, even a single episode of binge drinking, can cause serious damage. Reducing alcohol consumption or absorption is a direct way to alleviate the related harm. Alcohol is decomposed successively by alcohol dehydrogenase (ADH) and acetaldehyde dehydrogenase (ALDH) in the liver. Here, we produced a human ADH1B (hADH1B)-expressing probiotic, a recombinant Lactococcus lactis, that aimed to enhance alcohol degradation in the intestinal tract after oral administration. Our results showed that the oral hADH1B-expressing probiotic reduced alcohol absorption, prolonged the alcohol tolerance time, and shortened the recovery time after acute alcohol challenge. More importantly, the liver and intestine were protected from acute injury caused by alcohol challenge. Therefore, the engineered probiotic has the potential to protect organ damage from alcohol consumption. Furthermore, this engineered probiotic may have beneficial effects on alcohol-related diseases such as alcoholic fatty liver disease. IMPORTANCE Alcohol plays an important role in medical treatment, culture, and social interaction. However, excessive alcohol consumption or improper alcohol intake patterns can lead to serious damage to health. Aiming to reduce the harm of alcohol consumption, we designed a recombinant probiotic expressing hADH1B. Our results showed that this recombinant probiotic can reduce alcohol absorption and protect the body from alcohol damage, including hangover, liver, and intestinal damage. Reducing alcohol damage is helpful to the health of people with difficulty in abstinence. The engineered probiotic may provide new strategies for treatment and prevention of the negative effects of alcohol, and it also has the potential for widespread application.
Collapse
Affiliation(s)
- Xiaoxiao Jiang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Chunlong Yan
- Agriculture College of Yanbian University, Yanji, Jilin, China
| | - Hanlin Zhang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Li Chen
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Rui Jiang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Kexin Zheng
- Institute of Infectious Disease, Ditan Hospital, Capital Medical University, Beijing, China
| | - Wanzhu Jin
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Huijuan Ma
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Xiaomeng Liu
- Department of Nutrition and Food Hygiene, College of Public Health, Xinxiang Medical University, Xinxiang, Henan, China
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, China
| | - Meng Dong
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
39
|
Mead EA, Wang Y, Patel S, Thekkumthala AP, Kepich R, Benn-Hirsch E, Lee V, Basaly A, Bergeson S, Siegelmann HT, Pietrzykowski AZ. miR-9 utilizes precursor pathways in adaptation to alcohol in mouse striatal neurons. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2023; 3:11323. [PMID: 38116240 PMCID: PMC10730111 DOI: 10.3389/adar.2023.11323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
microRNA-9 (miR-9) is one of the most abundant microRNAs in the mammalian brain, essential for its development and normal function. In neurons, it regulates the expression of several key molecules, ranging from ion channels to enzymes, to transcription factors broadly affecting the expression of many genes. The neuronal effects of alcohol, one of the most abused drugs in the world, seem to be at least partially dependent on regulating the expression of miR-9. We previously observed that molecular mechanisms of the development of alcohol tolerance are miR-9 dependent. Since a critical feature of alcohol action is temporal exposure to the drug, we decided to better understand the time dependence of alcohol regulation of miR-9 biogenesis and expression. We measured the effect of intoxicating concentration of alcohol (20 mM ethanol) on the expression of all major elements of miR-9 biogenesis: three pri-precursors (pri-mir-9-1, pri-mir-9-2, pri-mir-9-3), three pre-precursors (pre-mir-9-1, pre-mir-9-2, pre-mir-9-3), and two mature microRNAs: miR-9-5p and miR-9-3p, using digital PCR and RT-qPCR, and murine primary medium spiny neurons (MSN) cultures. We subjected the neurons to alcohol based on an exposure/withdrawal matrix of different exposure times (from 15 min to 24 h) followed by different withdrawal times (from 0 h to 24 h). We observed that a short exposure increased mature miR-9-5p expression, which was followed by a gradual decrease and subsequent increase of the expression, returning to pre-exposure levels within 24 h. Temporal changes of miR-9-3p expression were complementing miR-9-5p changes. Interestingly, an extended, continuous presence of the drug caused a similar pattern. These results suggest the presence of the adaptive mechanisms of miR-9 expression in the presence and absence of alcohol. Measurement of miR-9 pre- and pri-precursors showed further that the primary effect of alcohol on miR-9 is through the mir-9-2 precursor pathway with a smaller contribution of mir-9-1 and mir-9-3 precursors. Our results provide new insight into the adaptive mechanisms of neurons to alcohol exposure. It would be of interest to determine next which microRNA-based mechanisms are involved in a transition from the acute, intoxicating effects of alcohol to the chronic, addictive effects of the drug.
Collapse
Affiliation(s)
- Edward Andrew Mead
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Yongping Wang
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Sunali Patel
- Thermo Fisher Scientific Inc., Austin, TX, United States
| | - Austin P. Thekkumthala
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Rebecca Kepich
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Elizabeth Benn-Hirsch
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Victoria Lee
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Azra Basaly
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Susan Bergeson
- Department of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Hava T. Siegelmann
- Department of Machine Learning, Mohamed bin Zayed University of Artificial Intelligence, Abu Dhabi, United Arab Emirates
- Biologically Inspired Neural & Dynamical Systems Laboratory, The Manning College of Information and Computer Sciences, University of Massachusetts, Amherst, MA, United States
| | - Andrzej Zbigniew Pietrzykowski
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
40
|
Chandrashekar DV, Steinberg RA, Han D, Sumbria RK. Alcohol as a Modifiable Risk Factor for Alzheimer's Disease-Evidence from Experimental Studies. Int J Mol Sci 2023; 24:9492. [PMID: 37298443 PMCID: PMC10253673 DOI: 10.3390/ijms24119492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/16/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by cognitive impairment and memory loss. Epidemiological evidence suggests that heavy alcohol consumption aggravates AD pathology, whereas low alcohol intake may be protective. However, these observations have been inconsistent, and because of methodological discrepancies, the findings remain controversial. Alcohol-feeding studies in AD mice support the notion that high alcohol intake promotes AD, while also hinting that low alcohol doses may be protective against AD. Chronic alcohol feeding to AD mice that delivers alcohol doses sufficient to cause liver injury largely promotes and accelerates AD pathology. The mechanisms by which alcohol can modulate cerebral AD pathology include Toll-like receptors, protein kinase-B (Akt)/mammalian target of rapamycin (mTOR) pathway, cyclic adenosine monophosphate (cAMP) response element-binding protein phosphorylation pathway, glycogen synthase kinase 3-β, cyclin-dependent kinase-5, insulin-like growth factor type-1 receptor, modulation of β-amyloid (Aβ) synthesis and clearance, microglial mediated, and brain endothelial alterations. Besides these brain-centric pathways, alcohol-mediated liver injury may significantly affect brain Aβ levels through alterations in the peripheral-to-central Aβ homeostasis. This article reviews published experimental studies (cell culture and AD rodent models) to summarize the scientific evidence and probable mechanisms (both cerebral and hepatic) by which alcohol promotes or protects against AD progression.
Collapse
Affiliation(s)
- Devaraj V. Chandrashekar
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618, USA;
| | - Ross A. Steinberg
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA 91711, USA; (R.A.S.); (D.H.)
| | - Derick Han
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA 91711, USA; (R.A.S.); (D.H.)
| | - Rachita K. Sumbria
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618, USA;
- Department of Neurology, University of California, Irvine, CA 92697, USA
| |
Collapse
|
41
|
Burgunder JM. Mechanisms underlying phenotypic variation in neurogenetic disorders. Nat Rev Neurol 2023:10.1038/s41582-023-00811-4. [PMID: 37202496 DOI: 10.1038/s41582-023-00811-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2023] [Indexed: 05/20/2023]
Abstract
Neurological diseases associated with pathogenic variants in a specific gene, or even with a specific pathogenic variant, can show profound phenotypic variation with regard to symptom presentation, age at onset and disease course. Highlighting examples from a range of neurogenetic disorders, this Review explores emerging mechanisms that are involved in this variability, including environmental, genetic and epigenetic factors that influence the expressivity and penetrance of pathogenic variants. Environmental factors, some of which can potentially be modified to prevent disease, include trauma, stress and metabolic changes. Dynamic patterns of pathogenic variants might explain some of the phenotypic variations, for example, in the case of disorders caused by DNA repeat expansions such as Huntington disease (HD). An important role for modifier genes has also been identified in some neurogenetic disorders, including HD, spinocerebellar ataxia and X-linked dystonia-parkinsonism. In other disorders, such as spastic paraplegia, the basis for most of the phenotypic variability remains unclear. Epigenetic factors have been implicated in disorders such as SGCE-related myoclonus-dystonia and HD. Knowledge of the mechanisms underlying phenotypic variation is already starting to influence management strategies and clinical trials for neurogenetic disorders.
Collapse
|
42
|
Rodrigues ADN, da Silva DCB, Baia-da-Silva DC, Mendes PFS, Ferreira MKM, Rocha GS, Freire MAM, Fernandes LMP, Maia CDSF, Gomes-Leal W, Lima RR. Aerobic Physical Training Attenuates Oxidative Stress in the Spinal Cord of Adult Rats Induced by Binge-like Ethanol Intake. Antioxidants (Basel) 2023; 12:antiox12051051. [PMID: 37237917 DOI: 10.3390/antiox12051051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/15/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Binge drinking is the most frequent consumption pattern among young adults and remarkably changes the central nervous system; thus, research on strategies to protect it is relevant. This study aimed to investigate the detrimental effects of binge-like EtOH intake on the spinal cord of male rats and the potential neuroprotective effects provided by moderate-intensity aerobic physical training. Male Wistar rats were distributed into the 'control group', 'training group', 'EtOH group', and 'training + EtOH'. The physical training protocol consisted of daily 30-min exercise on a treadmill for 5 consecutive days followed by 2 days off during 4 weeks. After the fifth day of each week, distilled water ('control group' and 'training group') or 3 g/kg of EtOH diluted at 20% w/v ('EtOH group' and 'training + EtOH group') was administered for 3 consecutive days through intragastric gavage to simulate compulsive consumption. Spinal cord samples were collected for oxidative biochemistry and morphometric analyses. The binge-like EtOH intake induced oxidative and tissue damage by decreasing reduced glutathione (GSH) levels, increasing lipid peroxidation (LPO), and reducing motor neurons (MN) density in the cervical segment. Even under EtOH exposure, physical training maintained GSH levels, reduced LPO, and prevented MN reduction at the cervical segment. Physical training is a non-pharmacological strategy to neuroprotect the spinal cord against oxidative damage induced by binge-like EtOH intake.
Collapse
Affiliation(s)
- Amanda do Nascimento Rodrigues
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil
| | - Diane Cleydes Baia da Silva
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil
| | - Daiane Claydes Baia-da-Silva
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil
| | - Paulo Fernando Santos Mendes
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil
| | - Maria Karolina Martins Ferreira
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil
| | - Gabriel Sousa Rocha
- Graduate Program in Health and Society, Faculty of Health Sciences, State University of Rio Grande do Norte, Mossoró 59610-110, Brazil
| | - Marco Aurelio M Freire
- Graduate Program in Health and Society, Faculty of Health Sciences, State University of Rio Grande do Norte, Mossoró 59610-110, Brazil
| | - Luanna Melo Pereira Fernandes
- Department of Morphology and Physiological Sciences, Center of Sciences Biological and Health, State University of Pará, Belém 66087-662, Brazil
| | - Cristiane do Socorro Ferraz Maia
- Laboratory of Pharmacology of Inflammation and Behavior, Health Sciences Institute, Pharmacy College, Federal University of Pará, Belém 66075-900, Brazil
| | - Walace Gomes-Leal
- Laboratory of Experimental Neuroprotection and Neuroregeneration, Institute of Collective Health, Federal University of Western Pará, Santarém 68040-470, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil
| |
Collapse
|
43
|
Xie L, Rungratanawanich W, Yang Q, Tong G, Fu E, Lu S, Liu Y, Akbar M, Song BJ, Wang X. Therapeutic strategies of small molecules in the microbiota-gut-brain axis for alcohol use disorder. Drug Discov Today 2023; 28:103552. [PMID: 36907319 PMCID: PMC10298843 DOI: 10.1016/j.drudis.2023.103552] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 03/04/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023]
Abstract
The microbiota-gut-brain axis (MGBA) is important in maintaining the structure and function of the central nervous system (CNS) and is regulated by the CNS environment and signals from the peripheral tissues. However, the mechanism and function of the MGBA in alcohol use disorder (AUD) are still not completely understood. In this review, we investigate the underlying mechanisms involved in the onset of AUD and/or associated neuronal deficits and create a foundation for better treatment (and prevention) strategies. We summarize recent reports focusing on the alteration of the MGBA in AUD. Importantly, we highlight the properties of small-molecule short-chain fatty acids (SCFAs), neurotransmitters, hormones, and peptides in the MGBA and discusses their usage as therapeutic agents against AUD.
Collapse
Affiliation(s)
- Lushuang Xie
- Departments of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China
| | - Wiramon Rungratanawanich
- Section of Molecular Pharmacology and Toxicology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD 20892, USA
| | - Qiang Yang
- Hubei Provincial Key Lab for Quality and Safety of Traditional Chinese Medicine Health Food, Jing Brand Research Institute, Daye, Hubei 435100, China
| | - Guoqiang Tong
- Hubei Provincial Key Lab for Quality and Safety of Traditional Chinese Medicine Health Food, Jing Brand Research Institute, Daye, Hubei 435100, China
| | - Eric Fu
- Departments of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Shiguang Lu
- Hubei Provincial Key Lab for Quality and Safety of Traditional Chinese Medicine Health Food, Jing Brand Research Institute, Daye, Hubei 435100, China
| | - Yuancai Liu
- Hubei Provincial Key Lab for Quality and Safety of Traditional Chinese Medicine Health Food, Jing Brand Research Institute, Daye, Hubei 435100, China
| | - Mohammed Akbar
- Division of Neuroscience & Behavior, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD 20892, USA
| | - Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD 20892, USA.
| | - Xin Wang
- Departments of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
44
|
Dugan MP, Ferguson LB, Hertz NT, Chalkley RJ, Burlingame AL, Shokat KM, Parker PJ, Messing RO. Chemical Genetic Identification of PKC Epsilon Substrates in Mouse Brain. Mol Cell Proteomics 2023; 22:100522. [PMID: 36863607 PMCID: PMC10105488 DOI: 10.1016/j.mcpro.2023.100522] [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: 07/15/2022] [Revised: 01/25/2023] [Accepted: 02/27/2023] [Indexed: 03/04/2023] Open
Abstract
PKC epsilon (PKCε) plays important roles in behavioral responses to alcohol and in anxiety-like behavior in rodents, making it a potential drug target for reducing alcohol consumption and anxiety. Identifying signals downstream of PKCε could reveal additional targets and strategies for interfering with PKCε signaling. We used a chemical genetic screen combined with mass spectrometry to identify direct substrates of PKCε in mouse brain and validated findings for 39 of them using peptide arrays and in vitro kinase assays. Prioritizing substrates with several public databases such as LINCS-L1000, STRING, GeneFriends, and GeneMAINA predicted interactions between these putative substrates and PKCε and identified substrates associated with alcohol-related behaviors, actions of benzodiazepines, and chronic stress. The 39 substrates could be broadly classified in three functional categories: cytoskeletal regulation, morphogenesis, and synaptic function. These results provide a list of brain PKCε substrates, many of which are novel, for future investigation to determine the role of PKCε signaling in alcohol responses, anxiety, responses to stress, and other related behaviors.
Collapse
Affiliation(s)
- Michael P Dugan
- Department of Neuroscience, The University of Texas at Austin, Austin, Texas, USA
| | - Laura B Ferguson
- Department of Neuroscience, The University of Texas at Austin, Austin, Texas, USA
| | - Nicholas T Hertz
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute at the University of California San Francisco, San Francisco, California, USA; Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - Robert J Chalkley
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - Kevan M Shokat
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute at the University of California San Francisco, San Francisco, California, USA
| | - Peter J Parker
- The Francis Crick Institute, London, United Kingdom; School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Robert O Messing
- Department of Neuroscience, The University of Texas at Austin, Austin, Texas, USA.
| |
Collapse
|
45
|
Moreira-Júnior RE, Guimarães MADF, Etcheverria da Silva M, Maioli TU, Faria AMC, Brunialti-Godard AL. Animal model for high consumption and preference of ethanol and its interplay with high sugar and butter diet, behavior, and neuroimmune system. Front Nutr 2023; 10:1141655. [PMID: 37063320 PMCID: PMC10097969 DOI: 10.3389/fnut.2023.1141655] [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: 01/10/2023] [Accepted: 03/10/2023] [Indexed: 03/31/2023] Open
Abstract
Introduction Mechanisms that dictate the preference for ethanol and its addiction are not only restricted to the central nervous system (CNS). An increasing body of evidence has suggested that abusive ethanol consumption directly affects the immune system, which in turn interacts with the CNS, triggering neuronal responses and changes, resulting in dependence on the drug. It is known that neuroinflammation and greater immune system reactivity are observed in behavioral disorders and that these can regulate gene transcription. However, there is little information about these findings of the transcriptional profile of reward system genes in high consumption and alcohol preference. In this regard, there is a belief that, in the striatum, an integrating region of the brain reward system, the interaction of the immune response and the transcriptional profile of the Lrrk2 gene that is associated with loss of control and addiction to ethanol may influence the alcohol consumption and preference. Given this information, this study aimed to assess whether problematic alcohol consumption affects the transcriptional profile of the Lrrk2 gene, neuroinflammation, and behavior and whether these changes are interconnected. Methods An animal model developed by our research group has been used in which male C57BL/6 mice and knockouts for the Il6 and Nfat genes were subjected to a protocol of high fat and sugar diet intake and free choice of ethanol in the following stages: Stage 1 (T1)-Dietary treatment, for 8 weeks, in which the animals receive high-calorie diet, High Sugar and Butter (HSB group), or standard diet, American Institute of Nutrition 93-Growth (AIN93G group); and Stage 2 (T2)-Ethanol consumption, in which the animals are submitted, for 4 weeks, to alcohol within the free choice paradigm, being each of them divided into 10 groups, four groups continued with the same diet and in the other six the HSB diet is substituted by the AIN93G diet. Five groups had access to only water, while the five others had a free choice between water and a 10% ethanol solution. The weight of the animals was evaluated weekly and the consumption of water and ethanol daily. At the end of the 12-week experiment, anxiety-like behavior was evaluated by the light/dark box test; compulsive-like behavior by Marble burying, transcriptional regulation of genes Lrrk2, Tlr4, Nfat, Drd1, Drd2, Il6, Il1β, Il10, and iNOS by RT-qPCR; and inflammatory markers by flow cytometry. Animals that the diet was replaced had an ethanol high preference and consumption. Results and discussion We observed that high consumption and preference for ethanol resulted in (1) elevation of inflammatory cells in the brain, (2) upregulation of genes associated with cytokines (Il6 and Il1β) and pro-inflammatory signals (iNOS and Nfat), downregulation of anti-inflammatory cytokine (Il10), dopamine receptor (Drd2), and the Lrrk2 gene in the striatum, and (3) behavioral changes such as decreased anxiety-like behavior, and increased compulsive-like behavior. Our findings suggest that interactions between the immune system, behavior, and transcriptional profile of the Lrrk2 gene influence the ethanol preferential and abusive consumption.
Collapse
Affiliation(s)
- Renato Elias Moreira-Júnior
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Andrade de Freitas Guimarães
- Laboratório de Imunobiologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Miguel Etcheverria da Silva
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Tatiani Uceli Maioli
- Laboratório de Imunobiologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Maria Caetano Faria
- Laboratório de Imunobiologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Lúcia Brunialti-Godard
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
46
|
Cheng Q, Xie M, Li G, Xue W, Zeng L, Ma D. Bacteria-Loaded Gastro-Retention Oral Delivery System for Alcohol Abuse. ACS Biomater Sci Eng 2023; 9:1460-1471. [PMID: 36848648 DOI: 10.1021/acsbiomaterials.3c00009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Alcohol abuse is harmful to human health, and many strategies have been developed to retard this harm through protecting liver or activating relative enzymes. In this study, a new strategy of decreasing the alcohol absorption directly depending on the dealcoholization by the bacteria in the upper gastrointestinal (GI) tract was reported. To realize this, a bacteria-loaded gastro-retention oral delivery system with pore structure was constructed through emulsification/internal gelation, which could relieve acute alcohol intoxication in mice successfully. It was found that this bacteria-loaded system kept the above 30% suspension ratio in the simulated gastric fluid for 4 min, displayed good protection effect for the bacteria, and decreased the alcohol concentration from 50 to 30% below within 24 h in vitro. The in vivo imaging results demonstrated that it remained in the upper GI tract until 24 h and reduced 41.9% alcohol absorption. The mice with oral administration of the bacteria-loaded system were found with normal gait, smooth coat, and less liver damage. Although the intestinal flora distribution was influenced slightly during the oral administration, it could restore to normal levels only one day after stopping oral administration quickly, suggesting good biosafety. In conclusion, these results revealed that the bacteria-loaded gastro-retention oral delivery system might intake alcohol molecules rapidly and has huge potential in the treatment of alcohol abuse.
Collapse
Affiliation(s)
- Qikun Cheng
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
- Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, Zhejiang 310015, China
| | - Mingzhi Xie
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Guowei Li
- Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Lexiang Zeng
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen (Zhongshan) University, Guangzhou 510120, People's Republic of China
| | - Dong Ma
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
- Artificial Organs and Materials Engineering Research Center, Ministry of Education, Guangzhou 510632, China
| |
Collapse
|
47
|
Ray S, Sil S, Kannan M, Periyasamy P, Buch S. Role of the gut-brain axis in HIV and drug abuse-mediated neuroinflammation. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2023; 3:11092. [PMID: 38389809 PMCID: PMC10880759 DOI: 10.3389/adar.2023.11092] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/23/2023] [Indexed: 02/24/2024]
Abstract
Drug abuse and related disorders are a global public health crisis affecting millions, but to date, limited treatment options are available. Abused drugs include but are not limited to opioids, cocaine, nicotine, methamphetamine, and alcohol. Drug abuse and human immunodeficiency virus-1/acquired immune deficiency syndrome (HIV-1/AIDS) are inextricably linked. Extensive research has been done to understand the effect of prolonged drug use on neuronal signaling networks and gut microbiota. Recently, there has been rising interest in exploring the interactions between the central nervous system and the gut microbiome. This review summarizes the existing research that points toward the potential role of the gut microbiome in the pathogenesis of HIV-1-linked drug abuse and subsequent neuroinflammation and neurodegenerative disorders. Preclinical data about gut dysbiosis as a consequence of drug abuse in the context of HIV-1 has been discussed in detail, along with its implications in various neurodegenerative disorders. Understanding this interplay will help elucidate the etiology and progression of drug abuse-induced neurodegenerative disorders. This will consequently be beneficial in developing possible interventions and therapeutic options for these drug abuse-related disorders.
Collapse
Affiliation(s)
- Sudipta Ray
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Susmita Sil
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Muthukumar Kannan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
48
|
Tousley AR, Yeh PWL, Yeh HH. Precocious emergence of cognitive and synaptic dysfunction in 3xTg-AD mice exposed prenatally to ethanol. Alcohol 2023; 107:56-72. [PMID: 36038084 PMCID: PMC10183974 DOI: 10.1016/j.alcohol.2022.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/01/2022] [Accepted: 08/09/2022] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, affecting approximately 50 million people worldwide. Early life risk factors for AD, including prenatal exposures, remain underexplored. Exposure of the fetus to alcohol (ethanol) is not uncommon during pregnancy, and may result in physical, behavioral, and cognitive changes that are first detected during childhood but result in lifelong challenges. Whether or not prenatal ethanol exposure may contribute to Alzheimer's disease risk is not yet known. Here we exposed a mouse model of Alzheimer's disease (3xTg-AD), bearing three dementia-associated transgenes, presenilin1 (PS1M146V), human amyloid precursor protein (APPSwe), and human tau (TauP301S), to ethanol on gestational days 13.5-16.5 using an established binge-type maternal ethanol exposure paradigm. We sought to investigate whether prenatal ethanol exposure resulted in a precocious onset or increased severity of AD progression, or both. We found that a brief binge-type gestational exposure to ethanol during a period of peak neuronal migration to the developing cortex resulted in an earlier onset of spatial memory deficits and behavioral inflexibility in the progeny, as assessed by performance on the modified Barnes maze task. The observed cognitive changes coincided with alterations to both GABAergic and glutamatergic synaptic transmission in layer V/VI neurons, diminished GABAergic interneurons, and increased β-amyloid accumulation in the medial prefrontal cortex. These findings provide the first preclinical evidence for prenatal ethanol exposure as a potential factor for modifying the onset of AD-like behavioral dysfunction and set the groundwork for more comprehensive investigations into the underpinnings of AD-like cognitive changes in individuals with fetal alcohol spectrum disorders.
Collapse
Affiliation(s)
- Adelaide R Tousley
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States; MD-PhD Program, Geisel School of Medicine at Dartmouth; Integrative Neuroscience at Dartmouth Graduate Program, Hanover, NH, United States
| | - Pamela W L Yeh
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Hermes H Yeh
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States.
| |
Collapse
|
49
|
Downs AM, Catavero CM, Kasten MR, McElligott ZA. Tauopathy and alcohol consumption interact to alter locus coeruleus excitatory transmission and excitability in male and female mice. Alcohol 2023; 107:97-107. [PMID: 36150608 DOI: 10.1016/j.alcohol.2022.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/22/2022] [Accepted: 08/29/2022] [Indexed: 12/23/2022]
Abstract
Alcohol use disorder is a major public health concern in the United States. Recent work has suggested a link between chronic alcohol consumption and the development of tauopathy disorders, such as Alzheimer's disease and frontotemporal dementia. However, relatively little work has investigated changes in neural circuitry involved in both tauopathy disorders and alcohol use disorder. The locus coeruleus (LC) is the major noradrenergic nucleus in the brain and is one of the earliest sites to be affected by tau lesions. The LC is also implicated in the rewarding effects of ethanol and alcohol withdrawal. In this study we assessed effects of long-term ethanol consumption and tauopathy on the physiology of LC neurons. Male and female P301S mice, a humanized transgenic mouse model of tauopathy, underwent 16 weeks of intermittent access to 20% ethanol from 3 to 7 months of age. We observed higher total alcohol consumption in female mice regardless of genotype. Male P301S mice consumed more ethanol and had a greater preference for ethanol than wild-type (WT) males. At the end of the drinking study, LC function was assessed using ex vivo whole cell electrophysiology. We found significant changes in excitatory inputs to the LC due to both ethanol and genotype. We found significantly increased excitability of the LC due to ethanol with greater effects in female P301S mice than in female WT mice. Our study identifies significant changes in the LC due to interactions between tauopathy and long-term ethanol use. These findings could have important implications regarding LC activity and changes in behavior due to both ethanol- and tauopathy-related dementia.
Collapse
Affiliation(s)
- Anthony M Downs
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Christina M Catavero
- Graduate Program in Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Michael R Kasten
- Department of Otolaryngology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Zoé A McElligott
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| |
Collapse
|
50
|
Yang C, Liao C, Zhao J, Guan Q, Wang G, Han Q. Dysregulation of tryptophan metabolism and distortion of cell signaling after oral exposure to ethanol and Kynurenic acid. Gene 2023; 852:147061. [PMID: 36423775 DOI: 10.1016/j.gene.2022.147061] [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: 08/29/2022] [Revised: 10/28/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022]
Abstract
Kynurenic acid (KYNA), an unavoidable tryptophan metabolite during fermentation is naturally blended with alcohol in all alcoholic beverages. Thus, alcohol drinking inevitably results in co-intake of KYNA. Effects of alcohol or KYNA on human health have been widely studied. However, the combined effects of both remain unknown. Here we report that alcohol and KYNA have a synergistic impact of on global gene expression, especially the gene sets related to tryptophan metabolism and cell signaling. Adult mice were exposed to alcohol (ethanol) and/or KYNA daily for a week. Transcriptomes of the brain, kidney and liver were profiled via bulk RNA sequencing. Results indicate that while KYNA alone largely promotes, and alcohol alone mostly inhibits gene expression, alcohol and KYNA co-administration has a stronger inhibition of global gene expression. Tryptophan metabolism is severely skewed towards kynurenine pathway by decreasing tryptophan hydroxylase 2 and increasing tryptophan dioxygenase. Quantification of tryptophan metabolic enzymes corroborates the transcriptional changes of these enzymes. Furthermore, the co-administration greatly enhances the GnRH signaling pathway. This research provides critical data to better understand the effects of alcohol and KYNA in mix on human health.
Collapse
Affiliation(s)
- Cihan Yang
- Laboratory of Tropical Veterinary Medicine and Vector Biology, School of Life Sciences, Hainan University, Haikou, Hainan 570228, China.
| | - Chenghong Liao
- Laboratory of Tropical Veterinary Medicine and Vector Biology, School of Life Sciences, Hainan University, Haikou, Hainan 570228, China; One Health Institute, Hainan University, Haikou, Hainan 570228, China.
| | - Jianguo Zhao
- Laboratory of Tropical Veterinary Medicine and Vector Biology, School of Life Sciences, Hainan University, Haikou, Hainan 570228, China; One Health Institute, Hainan University, Haikou, Hainan 570228, China.
| | - Qingfeng Guan
- Laboratory of Tropical Veterinary Medicine and Vector Biology, School of Life Sciences, Hainan University, Haikou, Hainan 570228, China; One Health Institute, Hainan University, Haikou, Hainan 570228, China.
| | - Guoshun Wang
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| | - Qian Han
- Laboratory of Tropical Veterinary Medicine and Vector Biology, School of Life Sciences, Hainan University, Haikou, Hainan 570228, China; One Health Institute, Hainan University, Haikou, Hainan 570228, China.
| |
Collapse
|