1
|
Li C, Wu Z, Chen F, Dai C, Yang X, Ye S, Shi M, Chen P, Liu X, Liu F. Regulation of Nrf2/GPX4 Signaling Pathway by Hyperbaric Oxygen Protects Against Depressive Behavior and Cognitive Impairment in a Spinal Cord Injury Rat Model. CNS Neurosci Ther 2025; 31:e70421. [PMID: 40326167 PMCID: PMC12053091 DOI: 10.1111/cns.70421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/20/2025] [Accepted: 04/23/2025] [Indexed: 05/07/2025] Open
Abstract
AIMS Neuroinflammation, microglial activation, and oxidative stress contribute to neuropsychiatric deficits following spinal cord injury (SCI). Hyperbaric oxygen (HBO) therapy has demonstrated anti-inflammatory, antioxidant, and neuroprotective properties. This study aimed to investigate the therapeutic effects and underlying mechanisms of HBO on depressive-like behavior, cognitive impairment, and hippocampal pathology in a rat model of SCI. METHODS We employed a battery of behavioral assessments, unbiased stereological analysis, immunofluorescence staining, and biochemical assays to evaluate neuroinflammation, oxidative stress, mitochondrial damage, and iron accumulation in the hippocampus. Untargeted proteomic analysis was conducted to identify potential molecular targets of HBO. Western blotting was used to assess the activation of the Nrf2/GPX4 signaling pathway. ML385, a selective Nrf2 inhibitor, was intrathecally administered 30 min prior to daily HBO treatment to validate pathway involvement. RESULTS HBO treatment significantly alleviated depressive-like behavior and cognitive deficits in SCI rats. It also suppressed M1-type microglial activation and reduced hippocampal neuroinflammation. Additionally, HBO mitigated neuronal ferroptosis induced by SCI through activation of the Nrf2/GPX4 signaling pathway. The protective effects of HBO were abolished by coadministration of ML385, confirming the critical role of Nrf2 signaling in mediating its anti-ferroptosis effects. CONCLUSION These findings highlight ferroptosis as a key pathological mechanism in SCI-induced hippocampal damage and suggest that HBO therapy alleviates depressive-like behavior and cognitive impairment by targeting the Nrf2/GPX4 pathway. This study provides new insights into the therapeutic potential of HBO in managing SCI-related neuropsychiatric dysfunction.
Collapse
Affiliation(s)
- Chenlu Li
- Department of Hyperbaric OxygenNational Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative DiseasesThe School of Basic Medical Sciences, Fujian Medical UniversityFuzhouFujianChina
| | - Zhongyue Wu
- Department of Hyperbaric OxygenNational Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
- Department of RadiologyFirst Affiliated Hospital of Fujian Medical UniversityFuzhouFujian ProvinceChina
| | - Fuxiang Chen
- Department of NeurosurgeryNeurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical UniversityFuzhouFujianChina
| | - Chaoxian Dai
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative DiseasesThe School of Basic Medical Sciences, Fujian Medical UniversityFuzhouFujianChina
| | - Xinyi Yang
- Department of Hyperbaric OxygenNational Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
- Department of RadiologyFirst Affiliated Hospital of Fujian Medical UniversityFuzhouFujian ProvinceChina
| | - Shumin Ye
- Department of Medicinal ChemistrySchool of Pharmacy, Fujian Medical UniversityFuzhouFujianChina
| | - Meng Shi
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative DiseasesThe School of Basic Medical Sciences, Fujian Medical UniversityFuzhouFujianChina
| | - Peng Chen
- Department of Sports MedicineNational Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical UniversityFujianChina
| | - Xueyan Liu
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative DiseasesThe School of Basic Medical Sciences, Fujian Medical UniversityFuzhouFujianChina
- Department of Medicinal ChemistrySchool of Pharmacy, Fujian Medical UniversityFuzhouFujianChina
| | - Fang Liu
- Department of Hyperbaric OxygenNational Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
| |
Collapse
|
2
|
Suloh H, Ojha SK, Kartawy M, Hamoudi W, Tripathi MK, Bazbaz W, Schottlender N, Ashery U, Khaliulin I, Amal H. Shared early molecular mechanisms revealed in P301S and 5xFAD Alzheimer's disease mouse models. Transl Psychiatry 2025; 15:97. [PMID: 40140365 PMCID: PMC11947184 DOI: 10.1038/s41398-025-03321-7] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 02/21/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder characterized by early molecular events that influence disease progression. Still, the molecular mechanisms caused by different mutations of AD are not understood. We have performed a multidisciplinary study to investigate and compare the early stages of the pathology in two transgenic AD mouse models: P301S and 5xFAD. Using SNOTRAP-based mass spectrometry, we assessed changes in S-nitrosylation, a nitric oxide-mediated post-translational modification, of proteins in both models during their juvenile age. The increased levels of 3-nitrotyrosine confirmed nitrosative stress in the mutant mice. Systems biology analysis revealed shared processes between the models, particularly in the γ-aminobutyric acid (GABA)ergic and glutamatergic neurotransmission processes. In the P301S model, we identified 273 S-nitrosylated (SNOed) proteins in the cortex, with 244 proteins uniquely SNOed in the diseased mice. In the 5xFAD model, 309 SNOed proteins were identified. We have found altered proteins expression of different glutamate/GABA-related markers in the cortex and hippocampus of both AD mouse models. Additionally, the phosphorylation levels of the mTOR signaling components revealed hyperactivation of this pathway in P301S mice. Conversely, 5xFAD mice showed no significant changes in mTOR signaling except for elevated phosphorylation of the ribosomal protein S6 in the cortex. Our findings revealed key molecular mechanisms in the two AD mouse models during their early stages. These mechanisms could serve as potential biomarkers and therapeutic targets for early-stage AD.
Collapse
Affiliation(s)
- Huda Suloh
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shashank Kumar Ojha
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maryam Kartawy
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Manish Kumar Tripathi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wisam Bazbaz
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nofar Schottlender
- School of Neurobiology, Biochemistry and Biophysics, Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Uri Ashery
- School of Neurobiology, Biochemistry and Biophysics, Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts; Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Li B, Li H, Chen H, Sui Y, Zeng J, Lin X, Fan Q, Song Z. Utilizing Hyperbaric Oxygen Therapy to Improve Cognitive Function in Patients With Alzheimer's Disease by Activating Autophagy-Related Signaling Pathways. Physiol Res 2025; 74:141-147. [PMID: 40126150 PMCID: PMC11995944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/03/2024] [Indexed: 03/25/2025] Open
Abstract
To investigate the impact of hyperbaric oxygen therapy (HBOT) on the cognitive function of mice with Alzheimer's disease (AD), while also identifying the cellular pathways associated with autophagy involved in the treatment. Twenty-four APP/PSl double transgenic mice were randomly assigned to either Group A or Group B, while another 24 C57 mice were randomly allocated to Group C or Group D. HBOT was administered to mice in Group B and Group D, and the Morris water maze test was used to assess changes in mice behavior. Histological examination using hematoxylin and eosin staining was conducted to observe pathological alterations in the hippocampus of the mice brain tissue. Polymerase chain reaction (PCR) was employed to analyze autophagy-related gene pathways in the hippocampus of the mice. Following HBOT, mice in Group B exhibited a significant reduction in escape latency and a notable increase in residence time within the target quadrant compared with Group A (P<0.05), as well as Group C and Group D (P<0.01). The hippocampal neurons in Group A and Group B mice exhibited disorganized arrangements, characterized by pyknosis and margination. Conversely, neurons in Group C displayed orderly arrangements, retaining intact structures with round nuclei demonstrating clear nuclear staining and normal morphology. The cellular morphology of mice in Group D remained unaffected. PCR analysis revealed no notable disparity in autophagy-related gene expression between Group A and Group C. However, the expression levels of five genes including Tgfb1, Mapk14, Bid, Atg7, and Akt1, were significantly elevated in Group B compared to Group A. HBOT has the potential to improve the cognitive function in mice modeled with AD. This improvement of cognitive function appears to be mediated by the up-regulation of autophagy-related genes, specifically Tgfb1, Mapk14, Bid, Atg7, and Akt1. These results indicate that HBOT may offer a therapeutic strategy for treating AD by enhancing autophagy mechanisms. Key words Alzheimer's disease, Autophagy, Hyperbaric oxygen, Morris water maze, PCR.
Collapse
Affiliation(s)
- B Li
- Department of Rehabilitation Medicine, The Affiliated Haikou Hospital of Xiangya Medical College, Central South University, No. 43 Renmin Avenue, Meilan District, Haikou 570208, Hainan, China. and
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Wang S, Xu H, Liu G, Chen L. Non-pharmacological treatment of Alzheimer's disease: an update. Front Aging Neurosci 2025; 17:1527242. [PMID: 40018518 PMCID: PMC11865074 DOI: 10.3389/fnagi.2025.1527242] [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: 11/13/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that significantly impairs memory, cognitive function, and the ability to perform daily tasks. The pathological features of AD include β-amyloid plaques, neurofibrillary tangles, and neuronal loss. Current AD treatments target pathological changes but often fail to noticeably slow disease progression and can cause severe complications, limiting their effectiveness. In addition to therapies targeting the core pathology of AD, a more comprehensive approach may be needed for its treatment. In recent years, non-pharmacological treatments such as physical therapy, exercise therapy, cell therapy, and nanoparticles have shown great potential in mitigating disease progression and alleviating clinical symptoms. This article reviews recent advances in non-pharmacological treatment approaches for AD, highlighting their contributions to AD management and facilitating the exploration of novel therapeutic strategies.
Collapse
Affiliation(s)
- Shaofen Wang
- West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, China
| | - Haochen Xu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Guangdong Liu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Limei Chen
- West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, China
| |
Collapse
|
5
|
Ho SY, Lin CH, Huang CC, Lin CH, Lin MT, Wang YJ, Ma JT, Shieh LT, Chang CP, Lin HJ. Hyperbaric oxygen therapy attenuates brain radiation-induced cognitive deficits in rats. Int J Med Sci 2025; 22:283-297. [PMID: 39781518 PMCID: PMC11704689 DOI: 10.7150/ijms.104248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/21/2024] [Indexed: 01/12/2025] Open
Abstract
Effective therapies for cognitive impairments induced by brain irradiation are currently lacking. This study investigated the therapeutic potential of hyperbaric oxygen therapy (HBOT) for radiation-induced brain injury in a randomized controlled experimental model using adult male Wistar rats. Adult male Wistar rats were divided into four experimental groups: 0 Gy whole brain radiotherapy (WBRT) with normal baric air (NBA) treatment, 0 Gy WBRT with HBOT, 10 Gy WBRT with NBA, and 10 Gy WBRT with HBOT. Behavioral tests and histochemical analyses were conducted four weeks post-WBRT to assess cognitive function, hippocampal microgliosis, apoptosis, and lipid peroxidation. Compared with the rats with 0 Gy WBRT on 28 days, the rats with 10 Gy WBRT on 28 days had significantly higher severity of spatial learning and memory dysfunction and hippocampal microgliosis, newborn neuronal apoptosis, and lipid peroxidation. HBOT significantly prevented and reversed WBRT-induced cognitive deficits, hippocampal microgliosis, newborn neuronal apoptosis, and lipid peroxidation. In addition, HBOT prevented and reversed the increased apoptosis among newborn neural stem cells and neuroblasts caused by 10 Gy WBRT on 7 days. The findings suggest that WBRT disrupts neurogenesis and enhance microgliosis, apoptosis of neuronal progenitors, and lipid peroxidation in the dentate gyrus, potentially leading to cognitive deficits and neuronal death. HBOT may offer a protective effect against these cognitive impairments and their underlying mechanisms in adult male rats following WBRT.
Collapse
Affiliation(s)
- Sheng-Yow Ho
- Department of Radiation Oncology, Chi Mei Medical Center, Tainan, Taiwan
- Graduate Institute of Medical Sciences, Chang Jung Christian University, Tainan, Taiwan
| | - Chia-Hui Lin
- Department of Radiation Oncology, Chi Mei Medical Center, Tainan, Taiwan
| | - Chien-Cheng Huang
- Department of Emergency Medicine, Chi Mei Medical Center, Tainan, Taiwan
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Emergency Medicine, Kaohsiung Medical University, Kaohsiung 81201, Taiwan
| | - Cheng-Hsien Lin
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Mao-Tsun Lin
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Ying-Jan Wang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jui-Ti Ma
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Li-Tsun Shieh
- Department of Radiation Oncology, Chi Mei Medical Center, Tainan, Taiwan
| | - Ching-Ping Chang
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Hung-Jung Lin
- Department of Emergency Medicine, Chi Mei Medical Center, Tainan, Taiwan
| |
Collapse
|
6
|
Choudhary S, Kumar V, Sharma K, Gour A, Sahrawat A, Jotshi A, Manhas D, Nandi U, Bharate SB, Ahmed Z, Kumar A. Crocetin Delays Brain and Body Aging by Increasing Cellular Energy Levels in Aged C57BL/6J Mice. ACS Pharmacol Transl Sci 2024; 7:3017-3033. [PMID: 39416964 PMCID: PMC11475333 DOI: 10.1021/acsptsci.4c00151] [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: 03/18/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 10/19/2024]
Abstract
Aging is usually accompanied by mitochondrial dysfunction, reduced energy levels, and cell death in the brain and other tissues. Mitochondria play a crucial role in maintaining cellular energy through oxidative phosphorylation (OXPHOS). However, OXPHOS is impaired as the mitochondrial oxygen supply decreases with age. We explored whether pharmacologically increased oxygen diffusion by crocetin can restore OXPHOS and help delay the aging of the brain and other vital organs. We found that aged mice treated with crocetin for four months displayed significantly improved memory behavior, neuromuscular coordination, and ATP and NAD+ levels in the brain and other vital organs, leading to an increased median life span. The transcriptomic analysis of hippocampi from crocetin-treated mice revealed that enhanced brain energy level was caused by the upregulation of genes linked to OXPHOS, and their expression was close to that in young mice. The chronic treatment of aged astrocytes also showed improved mitochondrial membrane potential and energy state of the cells. Moreover, chronic treatment with crocetin did not cause any oxidative stress. Our data suggest that restoring OXPHOS and the normal energy state of the cell can delay aging and enhance longevity. Therefore, molecules such as crocetin should be further explored to treat age-related diseases.
Collapse
Affiliation(s)
- Sushil Choudhary
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180016, India
- Academy
of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Vishnu Kumar
- Institute
of Anatomy and Cell Biology, Justus Liebig
University of Giessen, Giessen 35390, Germany
| | - Kuhu Sharma
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180016, India
- Academy
of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Abhishek Gour
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180016, India
- Academy
of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Ashish Sahrawat
- Molecular
Biophysics Unit, Indian Institute of Science, Bengaluru 560012, India
| | - Anshika Jotshi
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180016, India
- Academy
of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Diksha Manhas
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180016, India
- Academy
of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Utpal Nandi
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180016, India
- Academy
of Scientific and Innovative Research, Ghaziabad 201002, India
- Bose
Institute, Unified Academic Campus, Kolkata 700091, India
| | - Sandip B. Bharate
- Academy
of Scientific and Innovative Research, Ghaziabad 201002, India
- Natural Product
and Medicinal Chemistry Division, CSIR-Indian
Institute of Chemical Technology, Hyderabad 500007, India
| | - Zabeer Ahmed
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180016, India
- Academy
of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Ajay Kumar
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180016, India
- Academy
of Scientific and Innovative Research, Ghaziabad 201002, India
| |
Collapse
|
7
|
Bin-Alamer O, Abou-Al-Shaar H, Efrati S, Hadanny A, Beckman RL, Elamir M, Sussman E, Maroon JC. Hyperbaric oxygen therapy as a neuromodulatory technique: a review of the recent evidence. Front Neurol 2024; 15:1450134. [PMID: 39445195 PMCID: PMC11496187 DOI: 10.3389/fneur.2024.1450134] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/23/2024] [Indexed: 10/25/2024] Open
Abstract
Hyperbaric oxygen therapy (HBOT) has recently emerged as a promising neuromodulatory modality for treating several neurological and psychological disorders. Various studies indicate that HBOT can promote brain recovery and neuroplasticity through the modulation of key cellular and molecular mechanisms. HBOT affects multiple primary pathways and cellular functions including mitochondrial biogenesis and function (increased Bcl-2, reduced Bax, and enhanced ATP production), neurogenesis (upregulation of Wnt-3 and VEGF/ERK signaling), synaptogenesis (elevated GAP43 and synaptophysin expression), and anti-inflammatory responses (reduced TNF-α and IL-6). These mechanisms contribute to significant clinical benefits, such as enhanced cognitive function, improved recovery from traumatic brain injury and post-concussion syndrome, and symptom reduction in conditions like post-traumatic stress disorder and fibromyalgia. By influencing these molecular targets, HBOT offers a novel approach to neuromodulation that warrants further exploration. This review discusses the representative mechanisms of action of HBOT and highlights its therapeutic neuromodulatory effects and potential clinical applications across various neurological and psychiatric conditions.
Collapse
Affiliation(s)
- Othman Bin-Alamer
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Hussam Abou-Al-Shaar
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Shai Efrati
- Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf Harofeh) Medical Center, Be'er Ya'akov, Israel
| | - Amir Hadanny
- Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf Harofeh) Medical Center, Be'er Ya'akov, Israel
| | - Robert L. Beckman
- Foundation for the Study of Inflammatory Disease, Bethesda, MD, United States
| | | | | | - Joseph C. Maroon
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
8
|
Yu X, Zhao W, Liu Y, Lv J, Zhong X, Huang P. Hyperbaric oxygen therapy alleviates intestinal dysfunction following traumatic brain injury via m 6A regulation. Int J Med Sci 2024; 21:2272-2284. [PMID: 39310263 PMCID: PMC11413893 DOI: 10.7150/ijms.97682] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 08/04/2024] [Indexed: 09/25/2024] Open
Abstract
Hyperbaric oxygen (HBO) therapy can attenuate neurological impairment after traumatic brain injury (TBI) and alleviate intestinal dysfunction. However, the role and mechanism of HBO therapy in intestinal dysfunction following TBI remain unclear. Herein, by establishing a mouse model of controlled cortical impact (CCI), we found that HBO therapy reduced histopathological lesions and decreased the levels of inflammatory and oedema proteins in the intestinal tissues of mice 10 days after TBI. We also showed that HBO therapy improved microbiome abundance and probiotic (particularly g_Bifidobacterium) colonisation in mice post-CCI. Then, we identified that the m6A level imcreased notably in injured cortical tissue of CCI+HBO group compared with the CCI group following CCI. Thus, our results suggested that HBO therapy could alleviate TBI-induced intestinal dysfunction and m6A might participate in this regulation process, which provides new insights for exploring the specific mechanism and targets of HBO in the treatment of intestinal dysfunction after TBI, thereby improving the therapeutic effect of HBO.
Collapse
Affiliation(s)
- Xuelai Yu
- Department of Hyperbaric Oxygen, The Fourth Affiliated Hospital of Nanjing Medical University, 210031 Nanjing, China
| | - Wei Zhao
- Department of Neurosurgery, The Fourth Affiliated Hospital of Nanjing Medical University, 210031 Nanjing, China
| | - Yunyun Liu
- Department of Pathology, The Fourth Affiliated Hospital of Nanjing Medical University, 210031 Nanjing, China
| | - Jingchuan Lv
- Department of Intensive Care Unit, Nanjing Tongren Hospital, School of Medicine, Southeast University, 211102 Nanjing, China
| | - Xiang Zhong
- College of Animal Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Peizan Huang
- Department of Neurosurgery, The Fourth Affiliated Hospital of Nanjing Medical University, 210031 Nanjing, China
| |
Collapse
|
9
|
Choi YK. Detrimental Roles of Hypoxia-Inducible Factor-1α in Severe Hypoxic Brain Diseases. Int J Mol Sci 2024; 25:4465. [PMID: 38674050 PMCID: PMC11050730 DOI: 10.3390/ijms25084465] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Hypoxia stabilizes hypoxia-inducible factors (HIFs), facilitating adaptation to hypoxic conditions. Appropriate hypoxia is pivotal for neurovascular regeneration and immune cell mobilization. However, in central nervous system (CNS) injury, prolonged and severe hypoxia harms the brain by triggering neurovascular inflammation, oxidative stress, glial activation, vascular damage, mitochondrial dysfunction, and cell death. Diminished hypoxia in the brain improves cognitive function in individuals with CNS injuries. This review discusses the current evidence regarding the contribution of severe hypoxia to CNS injuries, with an emphasis on HIF-1α-mediated pathways. During severe hypoxia in the CNS, HIF-1α facilitates inflammasome formation, mitochondrial dysfunction, and cell death. This review presents the molecular mechanisms by which HIF-1α is involved in the pathogenesis of CNS injuries, such as stroke, traumatic brain injury, and Alzheimer's disease. Deciphering the molecular mechanisms of HIF-1α will contribute to the development of therapeutic strategies for severe hypoxic brain diseases.
Collapse
Affiliation(s)
- Yoon Kyung Choi
- Department of Integrative Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
10
|
Chiang MK, Lin TC, Lin KH, Chang YC, Hsieh-Li HM, Lai DM. Hyperbaric Oxygen Therapy Attenuated the Motor Coordination and Cognitive Impairment of Polyglutamine Spinocerebellar Ataxia SCA17 Mice. CEREBELLUM (LONDON, ENGLAND) 2024; 23:401-417. [PMID: 36943575 DOI: 10.1007/s12311-023-01548-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/14/2023] [Indexed: 03/23/2023]
Abstract
Spinocerebellar ataxias (SCAs) are a large and diverse group of autosomal-dominant neurodegenerative diseases. No drugs have been approved for these relentlessly progressive and fatal SCAs. Our previous studies indicate that oxidative stress, neuroinflammation, and neuronal apoptosis are elevated in the SCA17 mice, which are the main therapeutic targets of hyperbaric oxygen treatment (HBOT). HBOT is considered to be an alternative and less invasive therapy for SCAs. In this study, we evaluated the HBOT (2.2 ATA for 14 days) effect and the persistence for the management of SCA17 mice and their wild-type littermates. We found HBOT attenuated the motor coordination and cognitive impairment of SCA17 mice and which persisted for about 1 month after the treatment. The results of several biochemistry and liver/kidney hematoxylin and eosin staining show the HBOT condition has no obvious toxicity in the mice. Immunostaining analyses show that the neuroprotective effect of HBOT could be through the promotion of BDNF production and the amelioration of neuroinflammation. Surprisingly, HBOT executes different effects on the male and female SCA17 mice, including the reduction of neuroinflammation and activation of CaMKII and ERK. This study suggests HBOT is a potential alternative therapeutic treatment for SCA17. Accumulated findings have revealed the similarity in disease pathomechanisms and possible therapeutic strategies in polyQ diseases; therefore, HBOT could be an optional treatment as well as the other polyQ diseases.
Collapse
Affiliation(s)
- Meng-Ke Chiang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ta-Chun Lin
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | | | - Ya-Chin Chang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Hsiu Mei Hsieh-Li
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan.
| | - Dar-Ming Lai
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
11
|
Yang C, Liu G, Zeng X, Xiang Y, Chen X, Le W. Therapeutic effects of long-term HBOT on Alzheimer's disease neuropathologies and cognitive impairment in APP swe/PS1 dE9 mice. Redox Biol 2024; 70:103006. [PMID: 38241837 PMCID: PMC10831255 DOI: 10.1016/j.redox.2023.103006] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/01/2023] [Accepted: 12/18/2023] [Indexed: 01/21/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder with the pathological hallmarks of amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain. Although there is a hope that anti-amyloid monoclonal antibodies may emerge as a new therapy for AD, the high cost and side effect is a big concern. Non-drug therapy is attracting more attention and may provide a better resolution for the treatment of AD. Given the fact that hypoxia contributes to the pathogenesis of AD, hyperbaric oxygen therapy (HBOT) may be an effective intervention that can alleviate hypoxia and improve AD. However, it remains unclear whether long-term HBOT intervention in the early stage of AD can slow AD progression and ultimately prevent cognitive impairment in this disease. In this study we applied consecutive 3-month HBOT interventions on 3-month-old APPswe/PS1dE9 AD mice which represent the early stage of AD. When the APPswe/PS1dE9 mice at 9-month-old which represent the disease stage we measured cognitive function, 24-h blood oxygen saturation, Aβ and tau pathologies, vascular structure and function, and neuroinflammation in APPswe/PS1dE9 mice. Our results showed that long-term HBOT can attenuate the impairments in cognitive function observed in 9-month-old APPswe/PS1dE9 mice. Most importantly, HBOT effectively reduced the progression of Aβ plaques deposition, hyperphosphorylated tau protein aggregation, and neuronal and synaptic degeneration in the AD mice. Further, long-term HBOT was able to enhance blood oxygen saturation level. Besides, long-term HBOT can improve vascular structure and function, and reduce neuroinflammation in AD mice. This study is the first to demonstrate that long-term HBOT intervention in the early stage of AD can attenuate cognitive impairment and AD-like pathologies. Overall, these findings highlight the potential of long-term HBOT as a disease-modifying approach for AD treatment.
Collapse
Affiliation(s)
- Cui Yang
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Guangdong Liu
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Xianrong Zeng
- Department of Hyperbaric Oxygen, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yang Xiang
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Xi Chen
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Weidong Le
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China.
| |
Collapse
|
12
|
Shi R, Meng W, Liu Z, Chang L, Lu R, Chen X, Xue W, Deng Y. Hyperbaric oxygen therapy for poststroke insomnia: a systematic review and meta-analysis protocol. BMJ Open 2024; 14:e081642. [PMID: 38553058 PMCID: PMC10982781 DOI: 10.1136/bmjopen-2023-081642] [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: 11/02/2023] [Accepted: 03/13/2024] [Indexed: 04/02/2024] Open
Abstract
INTRODUCTION Insomnia stands as a frequent consequence of a cerebrovascular event, afflicting a substantial fraction of those who endure the aftermath of stroke. The ramifications of insomnia following a stroke can further exacerbate cognitive and behavioural anomalies while hindering the process of neurological convalescence. While several randomised controlled trials (RCTs) have scrutinised the effects of hyperbaric oxygen therapy (HBOT) on poststroke insomnia, the advantages and drawbacks persist in a state of ambiguity. We advocate for a systematic review and meta-analysis of randomised clinical trials to comprehensively evaluate the effectiveness and safety of HBOT in the context of poststroke insomnia. METHODS AND ANALYSIS A systematic search will be conducted from nine major databases (PubMed, Web of Science, EMBASE, VIP Information Database, Cochrane Central Register of Controlled Trials, China National Knowledge Infrastructure, China Biomedical Literature Database and Wanfang Database, Physiotherapy Evidence Database (PEDro)) for HBOT for poststroke insomnia of RCTs. The search procedures will adhere to a rigorous approach, commencing from the inception date of each database and continuing until 1 November 2023, with inquiries conducted exclusively in English and Chinese. The primary outcome will focus on the alteration in the quality of sleep while secondary outcomes will encompass the evaluation of adverse events and the rate of reoccurrence. The process of selecting studies, extracting data and evaluating the quality of research will be carried out independently by two reviewers. The quality of the included literature will be assessed using the tools of the Cochrane Collaboration. Meta-analysis will be performed by using RevMan V.5.4 and STATA V.16.0.b software. Finally, the quality of evidence will be assessed using the Grading of Recommendations, Assessment, Development and Evaluation method. ETHICS AND DISSEMINATION As all data are derived from published investigations via databases without direct patient contact, ethical approval is obviated in this study. The scientific studies will be published in professional academic publications. PROSPERO REGISTRATION NUMBER CRD42023468442.
Collapse
Affiliation(s)
- Rui Shi
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Wenyi Meng
- Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zhaozheng Liu
- Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Liping Chang
- Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Ruozhu Lu
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xingyu Chen
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Wen Xue
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yue Deng
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
13
|
Lin G, Zhao L, Lin J, Li X, Xu L. Clinical evidence of hyperbaric oxygen therapy for Alzheimer's disease: a systematic review and meta-analysis of randomized controlled trials. Front Aging Neurosci 2024; 16:1360148. [PMID: 38577491 PMCID: PMC10991696 DOI: 10.3389/fnagi.2024.1360148] [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: 12/28/2023] [Accepted: 03/06/2024] [Indexed: 04/06/2024] Open
Abstract
Objective To evaluate the potential benefits of hyperbaric oxygen intervention on people with Alzheimer's disease (AD) based on the existing randomized controlled trials (RCTs). Methods A systematic search was conducted in nine databases until November 17, 2023, for RCTs assessing the effect of hyperbaric oxygen intervention for AD. The primary outcomes included Mini-Mental State Examination (MMSE), Alzheimer's Disease Assessment Scale-Cognitive (ADAS-Cog), activities of daily living (ADL), and adverse events. All results were shown in forest plots, and sensitivity analysis was adopted to further verify the robustness of the pooled results. Results A total of 11 RCTs recruiting 847 participants were included in this meta-analysis. Based on the pooled evidence, hyperbaric oxygen could remarkably ameliorate MMSE [MD = 3.08, 95%CI (2.56, 3.61), p < 0.00001], ADAS-Cog [MD = -4.53, 95%CI (-5.05, -4.00), p < 0.00001], ADL [MD = 10.12, 95%CI (4.46, 15.79), p = 0.0005], MDA levels [SMD = -2.83, 95%CI (-5.27, -0.38), p = 0.02], SOD levels [SMD = 2.12, 95%CI (1.10, 3.15), p < 0.0001], IL-1-β levels [SMD = -1.00, 95%CI (-1.48, -0.53), p < 0.0001], and TGF-β1 levels [MD = 4.87, 95%CI (3.98, 5.76), p < 0.00001] without adverse events [OR = 1.17, 95%CI (0.68, 2.03), p = 0.58] for people with AD. The pooled results were robust after checking by sensitivity analysis. Conclusion These evidences suggest that hyperbaric oxygen is an effective and safe intervention for the treatment of AD. Further studies with more rigorous design will help to fully evaluate the clinical value of hyperbaric oxygen on cognition function in people with AD. Systematic review registration https://www.crd.york.ac.uk, identifier CRD42023483726.
Collapse
Affiliation(s)
| | | | | | | | - Lianwei Xu
- Department of Gynecology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
14
|
Balestra C, Baldelli S, Virgili F, Salvagno M, Mrakic-Sposta S, Fratantonio D. Pulsed Hyperoxia Acts on Plasmatic Advanced Glycation End Products and Advanced Oxidation Protein Products and Modulates Mitochondrial Biogenesis in Human Peripheral Blood Mononuclear Cells: A Pilot Study on the "Normobaric Oxygen Paradox". Int J Mol Sci 2024; 25:2394. [PMID: 38397071 PMCID: PMC10889761 DOI: 10.3390/ijms25042394] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/12/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
The "normobaric oxygen paradox" (NOP) describes the response to the return to normoxia after a hyperoxic event, sensed by tissues as an oxygen shortage, up-regulating redox-sensitive transcription factors. We have previously characterized the time trend of oxygen-sensitive transcription factors in human PBMCs, in which the return to normoxia after 30% oxygen is sensed as a hypoxic trigger, characterized by hypoxia-induced factor (HIF-1) activation. On the contrary, 100% and 140% oxygen induce a shift toward an oxidative stress response, characterized by NRF2 and NF-kB activation in the first 24 h post exposure. Herein, we investigate whether this paradigm triggers Advanced Glycation End products (AGEs) and Advanced Oxidation Protein Products (AOPPs) as circulating biomarkers of oxidative stress. Secondly, we studied if mitochondrial biogenesis was involved to link the cellular response to oxidative stress in human PBMCs. Our results show that AGEs and AOPPs increase in a different manner according to oxygen dose. Mitochondrial levels of peroxiredoxin (PRX3) supported the cellular response to oxidative stress and increased at 24 h after mild hyperoxia, MH (30% O2), and high hyperoxia, HH (100% O2), while during very high hyperoxia, VHH (140% O2), the activation was significantly high only at 3 h after oxygen exposure. Mitochondrial biogenesis was activated through nuclear translocation of PGC-1α in all the experimental conditions. However, the consequent release of nuclear Mitochondrial Transcription Factor A (TFAM) was observed only after MH exposure. Conversely, HH and VHH are associated with a progressive loss of NOP response in the ability to induce TFAM expression despite a nuclear translocation of PGC-1α also occurring in these conditions. This study confirms that pulsed high oxygen treatment elicits specific cellular responses, according to its partial pressure and time of administration, and further emphasizes the importance of targeting the use of oxygen to activate specific effects on the whole organism.
Collapse
Affiliation(s)
- Costantino Balestra
- Environmental, Occupational, Aging (Integrative) Physiology Laboratory, Haute Ecole Bruxelles-Brabant (HE2B), 1160 Brussels, Belgium
- Physical Activity Teaching Unit, Motor Sciences Department, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium
- DAN Europe Research Division (Roseto-Brussels), 1160 Brussels, Belgium
- Anatomical Research and Clinical Studies, Vrije Universiteit Brussels (VUB), 1090 Brussels, Belgium
| | - Sara Baldelli
- Department of Human Sciences and Promotion of the Quality of Life, IRCCS San Raffaele Pisana, San Raffaele Roma Open University, 00163 Rome, Italy
| | - Fabio Virgili
- Interuniversitary Consortium "National Institute for Bio-Structures and Bio-Systems"-I.N.B.B., 13, 00136 Rome, Italy
| | - Michele Salvagno
- Department of Intensive Care, Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium
| | - Simona Mrakic-Sposta
- Institute of Clinical Physiology, National Research Council (CNR), 20162 Milan, Italy
| | - Deborah Fratantonio
- Department of Medicine and Surgery, LUM University, S.S. 100 Km 18, 70100 Casamassima, Italy
| |
Collapse
|
15
|
Mensah-Kane P, Davis DL, Shi HS, Trinh OT, Vann PH, Dory L, Sumien N. Hyperbaric oxygen alleviates selective domains of cognitive and motor deficits in female 5xFAD mice. GeroScience 2024; 46:517-530. [PMID: 38153668 PMCID: PMC10828284 DOI: 10.1007/s11357-023-01047-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/14/2023] [Indexed: 12/29/2023] Open
Abstract
Treatment of Alzheimer's disease (AD) has been limited to managing of symptoms or anti-amyloid therapy with limited results and uncertainty. Seeking out new therapies that can reverse the effects of this devastating disease is important. Hyperbaric oxygen (HBO) therapy could be such a candidate as it has been shown to improve brain function in certain neurological conditions. Furthermore, the role sex plays in the vulnerability/resilience to AD remains equivocal. An understanding of what makes one sex more vulnerable to AD could unveil new pathways for therapy development. In this study, we investigated the effects of HBO on cognitive, motor, and affective function in a mouse model of AD (5xFAD) and assessed protein oxidation in peripheral tissues as a safety indicator. The motor and cognitive abilities of 5xFAD mice were significantly impaired. HBO therapy improved cognitive flexibility and associative learning of 5xFAD females but not males, but HBO had no effect other aspects of cognition. HBO also reversed AD-related declines in balance but had no impact on gait and anxiety-like behavior. HBO did not affect body weights or oxidative stress in peripheral tissues. Our study provides further support for HBO therapy as a potential treatment for AD and emphasizes the importance of considering sex as a biological variable in therapeutic development. Further investigations into the underlying mechanisms of HBO's sex-specific responses are warranted, as well as optimizing treatment protocols for maximum benefits.
Collapse
Affiliation(s)
- Paapa Mensah-Kane
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA
| | - Delaney L Davis
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA
| | - Helen S Shi
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA
| | - Oanh T Trinh
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA
| | - Philip H Vann
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA
| | - Ladislav Dory
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA
| | - Nathalie Sumien
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA.
| |
Collapse
|
16
|
Liu G, Yang C, Wang X, Chen X, Wang Y, Le W. Oxygen metabolism abnormality and Alzheimer's disease: An update. Redox Biol 2023; 68:102955. [PMID: 37956598 PMCID: PMC10665957 DOI: 10.1016/j.redox.2023.102955] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/13/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Oxygen metabolism abnormality plays a crucial role in the pathogenesis of Alzheimer's disease (AD) via several mechanisms, including hypoxia, oxidative stress, and mitochondrial dysfunction. Hypoxia condition usually results from living in a high-altitude habitat, cardiovascular and cerebrovascular diseases, and chronic obstructive sleep apnea. Chronic hypoxia has been identified as a significant risk factor for AD, showing an aggravation of various pathological components of AD, such as amyloid β-protein (Aβ) metabolism, tau phosphorylation, mitochondrial dysfunction, and neuroinflammation. It is known that hypoxia and excessive hyperoxia can both result in oxidative stress and mitochondrial dysfunction. Oxidative stress and mitochondrial dysfunction can increase Aβ and tau phosphorylation, and Aβ and tau proteins can lead to redox imbalance, thus forming a vicious cycle and exacerbating AD pathology. Hyperbaric oxygen therapy (HBOT) is a non-invasive intervention known for its capacity to significantly enhance cerebral oxygenation levels, which can significantly attenuate Aβ aggregation, tau phosphorylation, and neuroinflammation. However, further investigation is imperative to determine the optimal oxygen pressure, duration of exposure, and frequency of HBOT sessions. In this review, we explore the prospects of oxygen metabolism in AD, with the aim of enhancing our understanding of the underlying molecular mechanisms in AD. Current research aimed at attenuating abnormalities in oxygen metabolism holds promise for providing novel therapeutic approaches for AD.
Collapse
Affiliation(s)
- Guangdong Liu
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Cui Yang
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Xin Wang
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Xi Chen
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Yanjiang Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Weidong Le
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China; Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China.
| |
Collapse
|
17
|
Ehrenreich H, Gassmann M, Poustka L, Burtscher M, Hammermann P, Sirén AL, Nave KA, Miskowiak K. Exploiting moderate hypoxia to benefit patients with brain disease: Molecular mechanisms and translational research in progress. NEUROPROTECTION 2023; 1:9-19. [PMID: 37671067 PMCID: PMC7615021 DOI: 10.1002/nep3.15] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/17/2022] [Indexed: 09/07/2023]
Abstract
Hypoxia is increasingly recognized as an important physiological driving force. A specific transcriptional program, induced by a decrease in oxygen (O2) availability, for example, inspiratory hypoxia at high altitude, allows cells to adapt to lower O2 and limited energy metabolism. This transcriptional program is partly controlled by and partly independent of hypoxia-inducible factors. Remarkably, this same transcriptional program is stimulated in the brain by extensive motor-cognitive exercise, leading to a relative decrease in O2 supply, compared to the acutely augmented O2 requirement. We have coined the term "functional hypoxia" for this important demand-responsive, relative reduction in O2 availability. Functional hypoxia seems to be critical for enduring adaptation to higher physiological challenge that includes substantial "brain hardware upgrade," underlying advanced performance. Hypoxia-induced erythropoietin expression in the brain likely plays a decisive role in these processes, which can be imitated by recombinant human erythropoietin treatment. This article review presents hints of how inspiratory O2 manipulations can potentially contribute to enhanced brain function. It thereby provides the ground for exploiting moderate inspiratory plus functional hypoxia to treat individuals with brain disease. Finally, it sketches a planned multistep pilot study in healthy volunteers and first patients, about to start, aiming at improved performance upon motor-cognitive training under inspiratory hypoxia.
Collapse
Affiliation(s)
- Hannelore Ehrenreich
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Max Gassmann
- Institute of Veterinary Physiology and Zürich Center for Integrative Human Physiology, University of Zürich, Zürich, Switzerland
| | - Luise Poustka
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Martin Burtscher
- Faculty of Sports Science, University of Innsbruck, Innsbruck, Austria
| | | | - Anna-Leena Sirén
- Departments of Neurophysiology and Neurosurgery, University of Würzburg, Würzburg, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Kamilla Miskowiak
- Psychiatric Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
18
|
Dong Q, Wu M, Hu W. Efficacy and association of hyperbaric oxygen therapy combined with butylphthalide and oxiracetam with serum levels of inflammatory markers in vascular cognitive impairment after acute ischemic stroke. Pak J Med Sci 2023; 39:829-834. [PMID: 37250560 PMCID: PMC10214804 DOI: 10.12669/pjms.39.3.6828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/30/2022] [Accepted: 01/12/2022] [Indexed: 11/02/2023] Open
Abstract
Objectives This study evaluated the efficacy of hyperbaric oxygen therapy (HBOT) combined with butylphthalide (NBP) and oxiracetam (OXR) for vascular cognitive impairment after acute ischemic stroke and investigated the association between such combination therapy and the serum levels of inflammatory markers. Methods This was a prospective study which included eighty patients with post-AIS cognitive impairment (PAISCI) treated in Dongguan City People's Hospital from January 2020 to January 2022. They were randomized into study group and control group. The control group was provided with conventional therapy consisting of NBP for intravenous transfusion and oral OXR, while the study group received combination therapy of HBOT, NBP, and OXR. A comparison was drawn between the two groups regarding clinical outcomes, levels of recovery of cognitive and neurological function and intelligence, changes in inflammatory markers, and incidence of adverse drug reactions (ADRs). Results The response rate of the study group was significantly higher than that of the control group (p=0.04). The cognitive function scores of the study group were significantly better than those of the control group at the end of treatment (p<0.05). The post-treatment levels of inflammatory markers were significantly reduced in the study group when compared with the control group (p<0.05). At two weeks after treatment, the ADR rate of study group was significantly lower than the control group (p=0.03). Conclusions The combination therapy of HBOT, NBP, and OXR demonstrates robust efficacy in patients with PAISCI. It is deemed to be a safe and effective treatment regimen.
Collapse
Affiliation(s)
- Quanbin Dong
- Quanbin Dong, School of Health and Health, Dongguan Polytechnic, Dongguan 523000, Guangdong, China
| | - Mingxiu Wu
- Mingxiu Wu, Department of Gynecology and Obstetrics, Dongguan City People’s Hospital, Dongguan 523000, Guangdong, China
| | - Wenge Hu
- Wenge Hu, Department of Gynecology and Obstetrics, Dongguan Dongcheng People’s Hospital, Dongguan 523000, Guangdong, China
| |
Collapse
|
19
|
Mensah-Kane P, Sumien N. The potential of hyperbaric oxygen as a therapy for neurodegenerative diseases. GeroScience 2023; 45:747-756. [PMID: 36525211 PMCID: PMC9886764 DOI: 10.1007/s11357-022-00707-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 12/03/2022] [Indexed: 12/23/2022] Open
Abstract
The World Health Organization estimates that by the year 2040, neurodegenerative diseases will be the second leading cause of death in developed countries, overtaking cancer-related deaths and exceeded only by cardiovascular disease-related death. The search for interventions has therefore become paramount to alleviate some of this burden. Based on pathways affected in neurodegenerative diseases, hyperbaric oxygen treatment (HBOT) could be a good candidate. This therapy has been used for the past 50 years for conditions such as decompression sickness and wound healing and has been shown to have promising effects in conditions associated with neurodegeneration and functional impairments. The goal of this review was to explore the history of hyperbaric oxygen therapy, its uses, and benefits, and to evaluate its effectiveness as an intervention in treating neurodegenerative diseases. Additionally, we examined common mechanisms underlying the effects of HBOT in different neurodegenerative diseases, with a special emphasis on epigenetics.
Collapse
Affiliation(s)
- Paapa Mensah-Kane
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Nathalie Sumien
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA.
| |
Collapse
|
20
|
Carvalho C, Moreira PI. Metabolic defects shared by Alzheimer's disease and diabetes: A focus on mitochondria. Curr Opin Neurobiol 2023; 79:102694. [PMID: 36842275 DOI: 10.1016/j.conb.2023.102694] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 02/26/2023]
Abstract
Type 2 diabetes (T2D) and Alzheimer's disease (AD) are two global epidemics that share several metabolic defects, such as insulin resistance, impaired glucose metabolism, and mitochondrial defects. Importantly, strong evidence demonstrates that T2D significantly increases the risk of cognitive decline and dementia, particularly AD. Here, we provide an overview of the metabolic defects that characterize and link both pathologies putting the focus on mitochondria. The biomarker potential of mitochondrial components and the therapeutic potential of some drugs that target and modulate mitochondria are also briefly discussed.
Collapse
Affiliation(s)
- Cristina Carvalho
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Center for Innovation in Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
| | - Paula I Moreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Center for Innovation in Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
21
|
Le WD, Yang C, Yang Q, Xiang Y, Zeng XR, Xiao J. The neuroprotective effects of oxygen therapy in Alzheimer’s disease: a narrative review. Neural Regen Res 2023. [PMID: 35799509 PMCID: PMC9241400 DOI: 10.4103/1673-5374.343897] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Alzheimer’s disease (AD) is a degenerative neurological disease that primarily affects the elderly. Drug therapy is the main strategy for AD treatment, but current treatments suffer from poor efficacy and a number of side effects. Non-drug therapy is attracting more attention and may be a better strategy for treatment of AD. Hypoxia is one of the important factors that contribute to the pathogenesis of AD. Multiple cellular processes synergistically promote hypoxia, including aging, hypertension, diabetes, hypoxia/obstructive sleep apnea, obesity, and traumatic brain injury. Increasing evidence has shown that hypoxia may affect multiple pathological aspects of AD, such as amyloid-beta metabolism, tau phosphorylation, autophagy, neuroinflammation, oxidative stress, endoplasmic reticulum stress, and mitochondrial and synaptic dysfunction. Treatments targeting hypoxia may delay or mitigate the progression of AD. Numerous studies have shown that oxygen therapy could improve the risk factors and clinical symptoms of AD. Increasing evidence also suggests that oxygen therapy may improve many pathological aspects of AD including amyloid-beta metabolism, tau phosphorylation, neuroinflammation, neuronal apoptosis, oxidative stress, neurotrophic factors, mitochondrial function, cerebral blood volume, and protein synthesis. In this review, we summarized the effects of oxygen therapy on AD pathogenesis and the mechanisms underlying these alterations. We expect that this review can benefit future clinical applications and therapy strategies on oxygen therapy for AD.
Collapse
|
22
|
Ablin JN, Lang E, Catalogna M, Aloush V, Hadanny A, Doenyas-Barak K, Finci S, Polak N, Fishlev G, Korin C, Tzidky RY, Meir Genuth O, Efrati S. Hyperbaric oxygen therapy compared to pharmacological intervention in fibromyalgia patients following traumatic brain injury: A randomized, controlled trial. PLoS One 2023; 18:e0282406. [PMID: 36897850 PMCID: PMC10004612 DOI: 10.1371/journal.pone.0282406] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 02/08/2023] [Indexed: 03/11/2023] Open
Abstract
Fibromyalgia is a chronic pain syndrome with unsatisfactory response to current treatments. Physical trauma, including traumatic brain Injury (TBI) is among the etiological triggers. Hyperbaric Oxygen therapy (HBOT) is an intervention that combines 100% oxygen with elevated atmospheric pressure. HBOT has been applied as a neuro-modulatory treatment in central nervous system-related conditions. The current study investigated the utility of HBOT for TBI-related fibromyalgia. Fibromyalgia patients with a history of TBI were randomized to either HBOT or pharmacological intervention. HBOT protocol comprised 60 daily sessions, breathing 100% oxygen by mask at 2 absolute atmospheres (ATA) for 90 minutes. Pharmacological treatment included Pregabalin or Duloxetine. The primary outcome was subjective pain intensity on visual analogue scale (VAS); Secondary endpoints included questionnaires assessing fibromyalgia symptoms as well as Tc-99m-ECD SPECT brain imaging. Pain threshold and conditioned pain modulation (CPM) were also assessed. Results demonstrated a significant group-by-time interaction in pain intensity post-HBOT compared to the medication group (p = 0.001), with a large net effect size (d = -0.95) in pain intensity reduction following HBOT compared to medications. Fibromyalgia related symptoms and pain questionnaires demonstrated significant improvements induced by HBOT as well as improvements in quality of life and increase in pain thresholds and CPM. SPECT demonstrated significant group-by-time interactions between HBOT and medication groups in the left frontal and the right temporal cortex. In conclusion, HBOT can improve pain symptoms, quality of life, emotional and social function of patients suffering from FMS triggered by TBI. The beneficial clinical effect is correlated with increased brain activity in frontal and parietal regions, associated with executive function and emotional processing.
Collapse
Affiliation(s)
- Jacob N. Ablin
- Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Sackler School of Medicine, Tel- Aviv University, Tel-Aviv, Israel
- * E-mail:
| | - Erez Lang
- Sackler School of Medicine, Tel- Aviv University, Tel-Aviv, Israel
- Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf Harofeh) Medical Center, Be’er Ya’akov, Israel
| | - Merav Catalogna
- Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf Harofeh) Medical Center, Be’er Ya’akov, Israel
| | - Valerie Aloush
- Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Sackler School of Medicine, Tel- Aviv University, Tel-Aviv, Israel
| | - Amir Hadanny
- Sackler School of Medicine, Tel- Aviv University, Tel-Aviv, Israel
- Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf Harofeh) Medical Center, Be’er Ya’akov, Israel
| | - Keren Doenyas-Barak
- Sackler School of Medicine, Tel- Aviv University, Tel-Aviv, Israel
- Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf Harofeh) Medical Center, Be’er Ya’akov, Israel
| | - Shachar Finci
- Sackler School of Medicine, Tel- Aviv University, Tel-Aviv, Israel
- Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf Harofeh) Medical Center, Be’er Ya’akov, Israel
| | - Nir Polak
- Sackler School of Medicine, Tel- Aviv University, Tel-Aviv, Israel
- Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf Harofeh) Medical Center, Be’er Ya’akov, Israel
| | - Gregory Fishlev
- Sackler School of Medicine, Tel- Aviv University, Tel-Aviv, Israel
- Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf Harofeh) Medical Center, Be’er Ya’akov, Israel
| | - Calanit Korin
- Sackler School of Medicine, Tel- Aviv University, Tel-Aviv, Israel
- Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf Harofeh) Medical Center, Be’er Ya’akov, Israel
| | - Rachel Yehudit Tzidky
- Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf Harofeh) Medical Center, Be’er Ya’akov, Israel
| | - Oshra Meir Genuth
- Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf Harofeh) Medical Center, Be’er Ya’akov, Israel
| | - Shai Efrati
- Sackler School of Medicine, Tel- Aviv University, Tel-Aviv, Israel
- Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf Harofeh) Medical Center, Be’er Ya’akov, Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
23
|
Effect of Hyperbaric Oxygen Therapy on Sleep Quality, Drug Dosage, and Nerve Function in Patients with Sleep Disorders after Ischemic Cerebral Stroke. Emerg Med Int 2022. [DOI: 10.1155/2022/8307865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Objective. To explore the effects of hyperbaric oxygen therapy (HOT) on sleep quality, drug dosage, and nerve function in patients with sleep disorders after ischemic cerebral stroke (ICS). Methods. A total of 120 patients with acute ICS and sleep disorders who came to our hospital for treatment from January 2019 to October 2021 were selected and divided into control and observation groups according to the random numbering method, with 60 cases in each group. Both groups were treated with sertraline and eszopiclone for treating insomnia. The control group was given routine treatment for ICS, and the observation group was additionally treated with HOT in addition to the control group. The sleep quality, the use of sleep medication, the neurological function score, and the levels of serum tumor necrosis factor-α (TNF-α), endothelin (ET), and neuropeptide Y (NPY) before and after treatment were compared between the two groups. Results. The levels of TNF-α, ET and NPY were not significantly different between the two groups of patients before treatment (
), and all of the above indicators decreased significantly in both groups after treatment, with the observation group being lower than the control group (
). There was no significant difference in the sleep quality scores of PSQI, ESS, and SBQ between the two groups before treatment (
), and the above indicators decreased significantly in both groups after treatment, with the observation group being lower than the control group (
). There was no significant difference in the dose of sleep medication used in the first day of treatment between the two groups (
), and the amount of sleep medication used in the observation group was significantly less than that in the control group after 14 d of treatment (
). There was no significant difference in the NIHSS scores between the two groups before treatment (
), and the scores of both groups decreased after treatment, and the scores of the observation group were significantly lower than those of the control group (
). Conclusion. Compared with routine treatment, the addition of HOT to treat patients with sleep disorders after ICS can significantly improve their sleep quality, reduce dosage of sleep drugs, reduce inflammatory level of brain tissue and nerve function damage, and improve their prognosis. Trial Registration. This study was registered in the EA2019056
Collapse
|
24
|
The 2021 yearbook of Neurorestoratology. JOURNAL OF NEURORESTORATOLOGY 2022. [DOI: 10.1016/j.jnrt.2022.100008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
25
|
Fu Q, Duan R, Sun Y, Li Q. Hyperbaric oxygen therapy for healthy aging: From mechanisms to therapeutics. Redox Biol 2022; 53:102352. [PMID: 35649312 PMCID: PMC9156818 DOI: 10.1016/j.redox.2022.102352] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/17/2022] [Accepted: 05/23/2022] [Indexed: 12/19/2022] Open
Abstract
Hyperbaric oxygen therapy (HBOT), a technique through which 100% oxygen is provided at a pressure higher than 1 atm absolute (ATA), has become a well-established treatment modality for multiple conditions. The noninvasive nature, favorable safety profile, and common clinical application of HBOT make it a competitive candidate for several new indications, one of them being aging and age-related diseases. In fact, despite the conventional wisdom that excessive oxygen accelerates aging, appropriate HBOT protocols without exceeding the toxicity threshold have shown great promise in therapies against aging. For one thing, an extensive body of basic research has expanded our mechanistic understanding of HBOT. Interestingly, the therapeutic targets of HBOT overlap considerably with those of aging and age-related diseases. For another, pre-clinical and small-scale clinical investigations have provided validated information on the efficacy of HBOT against aging from various aspects. However, a generally applicable protocol for HBOT to be utilized in therapies against aging needs to be defined as a subsequent step. It is high time to look back and summarize the recent advances concerning biological mechanisms and therapeutic implications of HBOT in promoting healthy aging and shed light on prospective directions. Here we provide the first comprehensive overview of HBOT in the field of aging and geriatric research, which allows the scientific community to be aware of the emerging tendency and move beyond conventional wisdom to scientific findings of translational value.
Collapse
|
26
|
Saiyasit N, Butlig EAR, Chaney SD, Traylor MK, Hawley NA, Randall RB, Bobinger HV, Frizell CA, Trimm F, Crook ED, Lin M, Hill BD, Keller JL, Nelson AR. Neurovascular Dysfunction in Diverse Communities With Health Disparities-Contributions to Dementia and Alzheimer's Disease. Front Neurosci 2022; 16:915405. [PMID: 35844216 PMCID: PMC9279126 DOI: 10.3389/fnins.2022.915405] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease and related dementias (ADRD) are an expanding worldwide crisis. In the absence of scientific breakthroughs, the global prevalence of ADRD will continue to increase as more people are living longer. Racial or ethnic minority groups have an increased risk and incidence of ADRD and have often been neglected by the scientific research community. There is mounting evidence that vascular insults in the brain can initiate a series of biological events leading to neurodegeneration, cognitive impairment, and ADRD. We are a group of researchers interested in developing and expanding ADRD research, with an emphasis on vascular contributions to dementia, to serve our local diverse community. Toward this goal, the primary objective of this review was to investigate and better understand health disparities in Alabama and the contributions of the social determinants of health to those disparities, particularly in the context of vascular dysfunction in ADRD. Here, we explain the neurovascular dysfunction associated with Alzheimer's disease (AD) as well as the intrinsic and extrinsic risk factors contributing to dysfunction of the neurovascular unit (NVU). Next, we ascertain ethnoregional health disparities of individuals living in Alabama, as well as relevant vascular risk factors linked to AD. We also discuss current pharmaceutical and non-pharmaceutical treatment options for neurovascular dysfunction, mild cognitive impairment (MCI) and AD, including relevant studies and ongoing clinical trials. Overall, individuals in Alabama are adversely affected by social and structural determinants of health leading to health disparities, driven by rurality, ethnic minority status, and lower socioeconomic status (SES). In general, these communities have limited access to healthcare and healthy food and other amenities resulting in decreased opportunities for early diagnosis of and pharmaceutical treatments for ADRD. Although this review is focused on the current state of health disparities of ADRD patients in Alabama, future studies must include diversity of race, ethnicity, and region to best be able to treat all individuals affected by ADRD.
Collapse
Affiliation(s)
- Napatsorn Saiyasit
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Evan-Angelo R. Butlig
- Department of Neurology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Samantha D. Chaney
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Miranda K. Traylor
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Nanako A. Hawley
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Ryleigh B. Randall
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Hanna V. Bobinger
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Carl A. Frizell
- Department of Physician Assistant Studies, University of South Alabama, Mobile, AL, United States
| | - Franklin Trimm
- College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Errol D. Crook
- Department of Internal Medicine, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Mike Lin
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Benjamin D. Hill
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Joshua L. Keller
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Amy R. Nelson
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| |
Collapse
|
27
|
Hausman-Cohen S, Bilich C, Kapoor S, Maristany E, Stefani A, Wilcox A. Genomics as a Clinical Decision Support Tool for Identifying and Addressing Modifiable Causes of Cognitive Decline and Improving Outcomes: Proof of Concept Support for This Personalized Medicine Strategy. Front Aging Neurosci 2022; 14:862362. [PMID: 35517054 PMCID: PMC9062132 DOI: 10.3389/fnagi.2022.862362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/25/2022] [Indexed: 12/02/2022] Open
Abstract
The landscape of therapeutics for mild cognitive impairment and dementia is quite limited. While many single-agent trials of pharmaceuticals have been conducted, these trials have repeatedly been unable to show improvement in cognition. It is hypothesized that because Alzheimer’s, like many other chronic illnesses, is not a monogenic illness, but is instead caused by the downstream effects of an individual’s genetic variants interacting with each other, the environment, and lifestyle, that improving outcomes will require a personalized, precision medicine approach. This approach requires identifying and then addressing contributing genomic and other factors specific to each individual in a simultaneous fashion. Until recently, the utility of genomics as part of clinical decision-making for Alzheimer’s and cognitive decline has been limited by the lack of availability of a genomic platform designed specifically to evaluate factors contributing to cognitive decline and how to respond to these factors The clinical decision support (CDS) platform used in the cases presented focuses on common variants that relate to topics including, but not limited to brain inflammation, amyloid processing, nutrient carriers, brain ischemia, oxidative stress, and detoxification pathways. Potential interventions based on the scientific literature were included in the CDS, but the final decision on what interventions to apply were chosen by each patient’s physician. Interventions included supplements with “generally regarded as safe (GRAS)” rating, along with targeted diet and lifestyle modifications. We hypothesize that a personalized genomically targeted approach can improve outcomes for individuals with mild cognitive impairment who are at high risk of Alzheimer’s. The cases presented in this report represent a subset of cases from three physicians’ offices and are meant to provide initial proof of concept data demonstrating the efficacy of this method and provide support for this hypothesis. These patients were at elevated risk for Alzheimer’s due to their apolipoprotein E ε4 status. While further prospective and controlled trials need to be done, initial case reports are encouraging and lend support to this hypothesis of the benefit of a genomically targeted personalized medicine approach to improve outcomes in individuals with cognitive decline who are at high risk for Alzheimer’s.
Collapse
|
28
|
Hyperbaric Oxygen Treatment: Effects on Mitochondrial Function and Oxidative Stress. Biomolecules 2021; 11:biom11121827. [PMID: 34944468 PMCID: PMC8699286 DOI: 10.3390/biom11121827] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022] Open
Abstract
Hyperbaric oxygen treatment (HBOT)—the administration of 100% oxygen at atmospheric pressure (ATA) greater than 1 ATA—increases the proportion of dissolved oxygen in the blood five- to twenty-fold. This increase in accessible oxygen places the mitochondrion—the organelle that consumes most of the oxygen that we breathe—at the epicenter of HBOT’s effects. As the mitochondrion is also a major site for the production of reactive oxygen species (ROS), it is possible that HBOT will increase also oxidative stress. Depending on the conditions of the HBO treatment (duration, pressure, umber of treatments), short-term treatments have been shown to have deleterious effects on both mitochondrial activity and production of ROS. Long-term treatment, on the other hand, improves mitochondrial activity and leads to a decrease in ROS levels, partially due to the effects of HBOT, which increases antioxidant defense mechanisms. Many diseases and conditions are characterized by mitochondrial dysfunction and imbalance between ROS and antioxidant scavengers, suggesting potential therapeutic intervention for HBOT. In the present review, we will present current views on the effects of HBOT on mitochondrial function and oxidative stress, the interplay between them and the implications for several diseases.
Collapse
|
29
|
Gottfried I, Schottlender N, Ashery U. Hyperbaric Oxygen Treatment-From Mechanisms to Cognitive Improvement. Biomolecules 2021; 11:biom11101520. [PMID: 34680155 PMCID: PMC8533945 DOI: 10.3390/biom11101520] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/06/2021] [Accepted: 10/13/2021] [Indexed: 12/19/2022] Open
Abstract
Hyperbaric oxygen treatment (HBOT)—the medical use of oxygen at environmental pressure greater than one atmosphere absolute—is a very effective therapy for several approved clinical situations, such as carbon monoxide intoxication, incurable diabetes or radiation-injury wounds, and smoke inhalation. In recent years, it has also been used to improve cognition, neuro-wellness, and quality of life following brain trauma and stroke. This opens new avenues for the elderly, including the treatment of neurological and neurodegenerative diseases and improvement of cognition and brain metabolism in cases of mild cognitive impairment. Alongside its integration into clinics, basic research studies have elucidated HBOT’s mechanisms of action and its effects on cellular processes, transcription factors, mitochondrial function, oxidative stress, and inflammation. Therefore, HBOT is becoming a major player in 21st century research and clinical treatments. The following review will discuss the basic mechanisms of HBOT, and its effects on cellular processes, cognition, and brain disorders.
Collapse
Affiliation(s)
- Irit Gottfried
- School of Neurobiology, Biochemistry and Biophysics, Life Sciences Faculty, Tel Aviv University, Tel Aviv 6997801, Israel; (I.G.); (N.S.)
| | - Nofar Schottlender
- School of Neurobiology, Biochemistry and Biophysics, Life Sciences Faculty, Tel Aviv University, Tel Aviv 6997801, Israel; (I.G.); (N.S.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Uri Ashery
- School of Neurobiology, Biochemistry and Biophysics, Life Sciences Faculty, Tel Aviv University, Tel Aviv 6997801, Israel; (I.G.); (N.S.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Correspondence: ; Tel.: +972-3-6409827
| |
Collapse
|