1
|
Lee IT, Yang CC, Lin YJ, Wu WB, Lin WN, Lee CW, Tseng HC, Tsai FJ, Hsiao LD, Yang CM. Mevastatin-Induced HO-1 Expression in Cardiac Fibroblasts: A Strategy to Combat Cardiovascular Inflammation and Fibrosis. ENVIRONMENTAL TOXICOLOGY 2025; 40:264-280. [PMID: 39431643 DOI: 10.1002/tox.24429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/01/2024] [Accepted: 09/29/2024] [Indexed: 10/22/2024]
Abstract
Mevastatin (MVS) is known for its anti-inflammatory effects, potentially achieved by upregulating heme oxygenase-1 (HO-1), an enzyme involved in cytoprotection against oxidative injury. Nonetheless, the specific processes by which MVS stimulates HO-1 expression in human cardiac fibroblasts (HCFs) are not yet fully understood. In this study, we found that MVS treatment increased HO-1 mRNA and protein levels in HCFs. This induction was inhibited by pretreatment with specific inhibitors of p38 MAPK, JNK1/2, and FoxO1, and by siRNAs targeting NOX2, p47phox, p38, JNK1, FoxO1, Keap1, and Nrf2. MVS also triggered ROS generation and activated JNK1/2 and p38 MAPK, both attenuated by NADPH oxidase or ROS inhibitors. Additionally, MVS promoted the phosphorylation of FoxO1 and Nrf2, which was suppressed by p38 MAPK or JNK1/2 inhibitor. Furthermore, MVS inhibited TNF-α-induced NF-κB activation and vascular cell adhesion molecule-1 (VCAM-1) expression via the HO-1/CO pathway in HCFs. In summary, the induction of HO-1 expression in HCFs by MVS is mediated through two primary signaling pathways: NADPH oxidase/ROS/p38 MAPK, and JNK1/2/FoxO1 and Nrf2. This research illuminates the underlying processes through which MVS exerts its anti-inflammatory effects by modulating HO-1 in cardiac fibroblasts.
Collapse
Affiliation(s)
- I-Ta Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chien-Chung Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Taoyuan, Taoyuan, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yan-Jyun Lin
- Institute of Translational Medicine and new Drug Development, College of Medicine, China Medical University, Taichung, Taiwan
| | - Wen-Bin Wu
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Wei-Ning Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chiang-Wen Lee
- Department of Nursing, Division of Basic Medical Sciences, and Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Chiayi, Taiwan
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi, Taiwan
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Hui-Ching Tseng
- Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Fuu-Jen Tsai
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Division of Medical Genetics, China Medical University Children's Hospital, Taichung, Taiwan
| | - Li-Der Hsiao
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chuen-Mao Yang
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| |
Collapse
|
2
|
Özden ES, Özcan MS, Savran M, Ilhan I, Tepebası MY, Sevuk MA, Özmen Ö. Effects of Tasimelteon Treatment on Traumatic Brain Injury Through NRF-2/HO-1 and RIPK1/RIPK3/MLKL Pathways in Rats. Mol Neurobiol 2025:10.1007/s12035-025-04711-0. [PMID: 39878865 DOI: 10.1007/s12035-025-04711-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025]
Abstract
Secondary brain damageafter traumatic brain injury (TBI) involves oxidative stress, neuroinflammation, apoptosis, and necroptosis and can be reversed by understanding these molecular pathways. The objective of this study was to examine the impact of tasimelteon (Tasi) administration on brain injury through the nuclear factor erythroid 2-related factor 2 (NRF-2)/heme oxygenase-1 (HO-1) and receptor-interacting protein kinase 1 (RIPK1)/receptor-interacting protein kinase 3 (RIPK3)/mixed lineage kinase domain-like (MLKL) pathways in rats with TBI. Thirty-two male Wistar albino rats weighing 300-350 g were randomly divided into four groups: the control group, trauma group, Tasi-1 group (trauma + 1 mg/kg Tasi intraperitoneally), and Tasi-10 group (trauma + 10 mg/kg Tasi intraperitoneally). At the end of the experimental phase, after sacrifice, blood samples and brain tissue were collected for biochemical, histopathological, immunohistochemical, and genetic analyses. Tasi increased the total antioxidant status and decreased the total oxidant status and oxidative stress index. In addition, Tasi caused histopathological changes characterized by a markedly reduced hemorrhage area in the Tasi-1 group. Normal brain and meningeal structure was observed in rats in the Tasi-10 group. Immunohistochemical analysis indicated that Tasi also decreased the expression of interferon-gamma, caspase-3, and tumor necrosis factor-alpha in the brain tissue. Although NRF-2 and HO-1 expression decreased, RIPK1/RIPK3/MLKL gene expression increased due to trauma. However, Tasi treatment reversed all these findings. Tasi protected against brain injury through the NRF-2/HO-1 and RIPK1/RIPK3/MLKL pathways in rats with TBI.
Collapse
Affiliation(s)
- Eyyüp Sabri Özden
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Suleyman Demirel University, Cunur, 32260, Isparta, Turkey.
| | - Mustafa Soner Özcan
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Suleyman Demirel University, Cunur, 32260, Isparta, Turkey
| | - Mehtap Savran
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Ilter Ilhan
- Department of Biochemistry, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | | | - Mehmet Abdulkadir Sevuk
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Özlem Özmen
- Department of Pathology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| |
Collapse
|
3
|
Yang C, He Y, Ren S, Ding Y, Liu X, Li X, Sun H, Jiao D, Zhang H, Wang Y, Sun L. Hydrogen Attenuates Cognitive Impairment in Rat Models of Vascular Dementia by Inhibiting Oxidative Stress and NLRP3 Inflammasome Activation. Adv Healthc Mater 2024; 13:e2400400. [PMID: 38769944 DOI: 10.1002/adhm.202400400] [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: 02/02/2024] [Revised: 05/09/2024] [Indexed: 05/22/2024]
Abstract
Vascular dementia (VaD) is the second most common form of dementia worldwide. Oxidative stress and neuroinflammation are important factors contributing to cognitive dysfunction in patients with VaD. The antioxidant and anti-inflammatory properties of hydrogen are increasingly being utilized in neurological disorders, but conventional hydrogen delivery has the disadvantage of inefficiency. Therefore, magnesium silicide nanosheets (MSNs) are used to release hydrogen in vivo in larger quantities and for longer periods of time to explore the appropriate dosage and regimen. In this study, it is observed that hydrogen improved learning and working memory in VaD rats in the Morris water maze and Y-maze, which elicits improved cognitive function. Nissl staining of neurons shows that hydrogen treatment significantly improves edema in neuronal cells. The expression and activation of reactive oxygen species (ROS), Thioredoxin-interacting protein (TXNIP), NOD-like receptor protein 3 (NLRP3), caspase-1, and IL-1β in the hippocampus are measured via ELISA, Western blotting, real-time qPCR, and immunofluorescence. The results show that oxidative stress indicators and inflammasome-related factors are significantly decreased after 7dMSN treatment. Therefore, it is concluded that hydrogen can ameliorate neurological damage and cognitive dysfunction in VaD rats by inhibiting ROS/NLRP3/IL-1β-related oxidative stress and inflammation.
Collapse
Affiliation(s)
- Congwen Yang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Yuxuan He
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Shuang Ren
- Department of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Yiqin Ding
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Xinru Liu
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Xue Li
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Hao Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Dezhi Jiao
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Haolin Zhang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Yingshuai Wang
- Department of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Lin Sun
- School of Psychology, Shandong Second Medical University, Weifang, Shandong, 261053, China
| |
Collapse
|
4
|
Wu F, Liang T, Liu Y, Wang C, Sun Y, Wang B. Effects of perioperative hydrogen inhalation on brain edema and prognosis in patients with glioma: a single-center, randomized controlled study. Front Neurol 2024; 15:1413904. [PMID: 39099781 PMCID: PMC11294077 DOI: 10.3389/fneur.2024.1413904] [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: 04/08/2024] [Accepted: 07/09/2024] [Indexed: 08/06/2024] Open
Abstract
Introduction Brain edema is a life-threatening complication that occurs after glioma surgery. There are no noninvasive and specific treatment methods for brain edema. Hydrogen is an anti-inflammatory and antioxidant gas that has demonstrated therapeutic and preventative effects on several diseases, particularly in the nervous system. This study aimed to determine the therapeutic effects of hydrogen administration on brain edema following glioma surgery and elucidate its mechanism. Methods A single-center, randomized controlled clinical trial of hydrogen inhalation was conducted (China Clinical Trial Registry [ChiCTR-2300074362]). Participants in hydrogen (H) group that inhaled hydrogen experienced quicker alleviation of postoperative brain edema compared with participants in control (C) group that inhaled oxygen. Results The volume of brain edema before discharge was significantly lower in the H group (p < 0.05). Additionally, the regression rate of brain edema was higher in the H group than in the C group, which was statistically significant (p < 0.05). Furthermore, 3 days after surgery, the H group had longer total sleep duration, improved sleep efficiency, shorter sleep latency, and lower numerical rating scale (NRS) scores (p < 0.05). Discussion In conclusion, hydrogen/oxygen inhalation effectively reduced postoperative brain edema in glioma patients. Further research is necessary to understand the underlying mechanisms of hydrogen's therapeutic effects. Hydrogen is expected to become a new target for future adjuvant therapy for brain edema.
Collapse
Affiliation(s)
- Fan Wu
- Department of Anesthesiology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Tao Liang
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yang Liu
- Department of Anesthesiology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Chenhui Wang
- Department of Anesthesiology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Yongxing Sun
- Department of Anesthesiology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Baoguo Wang
- Department of Anesthesiology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Min Y, Yu ZQ. GSK'872 Improves Prognosis of Traumatic Brain Injury by Switching Receptor-Interacting Serine/Threonine-Protein Kinase 3-dependent Necroptosis to Cysteinyl Aspartate Specific Proteinase-8-Dependent Apoptosis. World Neurosurg 2024; 187:e136-e147. [PMID: 38636634 DOI: 10.1016/j.wneu.2024.04.051] [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: 11/27/2023] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Traumatic brain injury (TBI) is an important health concern in the society. Previous studies have suggested that necroptosis occurs following TBI. However, the underlying mechanisms and roles of necroptosis are not well understood. In this study, we aimed to assess the role of receptor-interacting serine/threonine-protein kinase 3 (RIP3)-mediated necroptosis after TBI both in vitro and in vivo. METHODS We established a cell-stretching injury and mouse TBI model by applying a cell injury controller and controlled cortical impactor to evaluate the relationships among necroptosis, apotosis, inflammation, and TBI both in vitro and in vivo. RESULTS The results revealed that necroptosis mediated by RIP1, RIP3, and mixed lineage kinase domain-like protein was involved in secondary TBI. Additionally, protein kinase B (Akt), phosphorylated Akt, mammalian target of rapamycin (mTOR), and phosphorylated mTOR potentially contribute to necroptosis. The inhibition of RIP3 by GSK'872 (a specific inhibitor) blocked necroptosis and reduced the activity of Akt/mTOR, leading to the alleviation of inflammation by reducing the levels of NOD-, LRR- and pyrin domain-containing protein 3. Moreover, the inhibition of RIP3 by GSK'872 promoted the activity of cysteinyl aspartate specific proteinase-8, an enzyme involved in apoptosis and inflammation. CONCLUSIONS These data demonstrate that RIP3 inhibition could improve the prognosis of TBI, based on the attenuation of inflammation by switching RIP3-dependent necroptosis to cysteinyl aspartate specific proteinase-8-dependent apoptosis.
Collapse
Affiliation(s)
- Yue Min
- Department of Neurosurgery, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Ze-Qi Yu
- Department of Neurosurgery, Armed Police Force Hospital of Sichuan, Leshan, Sichuan, China.
| |
Collapse
|
6
|
Feng Y, Wu C, Song B, Zhang Y, Jiang M, Qi Z, Chen L, Li A, Ye H, Liu B, Feng Y, Ji X, Ma Z, Li M. Investigation of neuroprotective effects of H 2 by CiteSpace-based bibliometric analysis. Brain Circ 2024; 10:229-239. [PMID: 39526111 PMCID: PMC11542759 DOI: 10.4103/bc.bc_111_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND AND AIMS Neuroprotection plays an important role in the treatment of brain disorders. In recent years, studies using rat models and clinical trials have demonstrated the positive effects of hydrogen treatment on neurological disorders and brain injuries. Hence, it is of great significance to shed light on this issue. In this article, CiteSpace is employed for visualization and bibliometric analysis of the research frontiers and evolving trends related to the neuroprotective effect of hydrogen. METHODS All articles published from 2009 to 2023 that discussed the neuroprotective effects of hydrogen in cerebrovascular diseases were retrieved from the Web of Science. Using CiteSpace, a visualization analysis was conducted on aspects such as countries, institutions, authors, keywords, and Co cited references, which enables an intuitive observation of current research hotspots. RESULTS After manual screening, a total of 106 articles were retrieved. Over time, The number of publications has increased annually. Regarding national contributions, the top three countries with the highest number of publications include China, the United States, and Japan. The Second Military Medical University is the institution that publishes the most articles and has significant influence in the field of hydrogen neuroprotection. Sun, Xuejun and Domoki, Ferenc were the most productive. The most common keywords include hydrogen, oxidative stress, inflammation, and apoptosis. Potential areas of focus for future research consist of early brain injury, hydrogen, ischemia-reperfusion injury and hypothermia treatment. CONCLUSION The bibliometric study presented herein offers insights into the current status and trends of research on hydrogen in the field of cerebrovascular diseases. Future research trends suggest that hydrogen contributes significantly to the cerebrovascular domain through its anti-inflammatory, antioxidative, and anti-apoptotic mechanisms. This study can aid researchers in identifying hot topics and exploring new research directions.
Collapse
Affiliation(s)
- Yan Feng
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Chuanjie Wu
- Department of Neurology, Beijing Institute of Geriatrics , Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Baoying Song
- Department of Neurology, Beijing Institute of Geriatrics , Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yang Zhang
- Department of Neurology, Beijing Institute of Geriatrics , Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Miaowen Jiang
- Department of Neurology, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Zhengfei Qi
- Department of Neurology, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Le Chen
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Anzhi Li
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hanming Ye
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Biluo Liu
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yu Feng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xunming Ji
- Department of Neurology, Beijing Institute of Geriatrics , Xuanwu Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Zhengfei Ma
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ming Li
- Department of Neurology, Beijing Institute of Geriatrics , Xuanwu Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Artamonov MY, LeBaron TW, Pyatakovich FA, Minenko IA. Mesenchymal Stem Cell Priming: Potential Benefits of Administration of Molecular Hydrogen. Pharmaceuticals (Basel) 2024; 17:469. [PMID: 38675429 PMCID: PMC11054387 DOI: 10.3390/ph17040469] [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: 10/25/2023] [Revised: 01/19/2024] [Accepted: 02/10/2024] [Indexed: 04/28/2024] Open
Abstract
Stem cell therapy has emerged as a promising avenue for regenerative medicine, offering the potential to treat a wide range of debilitating diseases and injuries. Among the various types of stem cells, mesenchymal stem cells (MSCs) have garnered significant attention due to their unique properties and therapeutic potential. In recent years, researchers have been exploring novel approaches to enhance the effectiveness of MSC-based therapies. One such approach that has gained traction is the priming of MSCs with molecular hydrogen (H2). This article delves into the fascinating world of mesenchymal stem cell priming with molecular hydrogen and the potential benefits it holds for regenerative medicine.
Collapse
Affiliation(s)
| | - Tyler W. LeBaron
- Department of Kinesiology and Outdoor Recreation, Southern Utah University, Cedar City, UT 84720, USA
- Molecular Hydrogen Institute, Enoch, UT 84721, USA
| | | | | |
Collapse
|
8
|
Silvestro S, Raffaele I, Quartarone A, Mazzon E. Innovative Insights into Traumatic Brain Injuries: Biomarkers and New Pharmacological Targets. Int J Mol Sci 2024; 25:2372. [PMID: 38397046 PMCID: PMC10889179 DOI: 10.3390/ijms25042372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/08/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
A traumatic brain injury (TBI) is a major health issue affecting many people across the world, causing significant morbidity and mortality. TBIs often have long-lasting effects, disrupting daily life and functionality. They cause two types of damage to the brain: primary and secondary. Secondary damage is particularly critical as it involves complex processes unfolding after the initial injury. These processes can lead to cell damage and death in the brain. Understanding how these processes damage the brain is crucial for finding new treatments. This review examines a wide range of literature from 2021 to 2023, focusing on biomarkers and molecular mechanisms in TBIs to pinpoint therapeutic advancements. Baseline levels of biomarkers, including neurofilament light chain (NF-L), ubiquitin carboxy-terminal hydrolase-L1 (UCH-L1), Tau, and glial fibrillary acidic protein (GFAP) in TBI, have demonstrated prognostic value for cognitive outcomes, laying the groundwork for personalized treatment strategies. In terms of pharmacological progress, the most promising approaches currently target neuroinflammation, oxidative stress, and apoptotic mechanisms. Agents that can modulate these pathways offer the potential to reduce a TBI's impact and aid in neurological rehabilitation. Future research is poised to refine these therapeutic approaches, potentially revolutionizing TBI treatment.
Collapse
Affiliation(s)
| | | | | | - Emanuela Mazzon
- IRCCS Centro Neurolesi Bonino Pulejo, Via Provinciale Palermo, SS 113, Contrada Casazza, 98124 Messina, Italy; (S.S.); (I.R.); (A.Q.)
| |
Collapse
|
9
|
Obara T, Naito H, Nojima T, Hirayama T, Hongo T, Ageta K, Aokage T, Hisamura M, Yumoto T, Nakao A. Hydrogen in Transplantation: Potential Applications and Therapeutic Implications. Biomedicines 2024; 12:118. [PMID: 38255223 PMCID: PMC10813693 DOI: 10.3390/biomedicines12010118] [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: 12/14/2023] [Revised: 12/25/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Hydrogen gas, renowned for its antioxidant properties, has emerged as a novel therapeutic agent with applications across various medical domains, positioning it as a potential adjunct therapy in transplantation. Beyond its antioxidative properties, hydrogen also exerts anti-inflammatory effects by modulating pro-inflammatory cytokines and signaling pathways. Furthermore, hydrogen's capacity to activate cytoprotective pathways bolsters cellular resilience against stressors. In recent decades, significant advancements have been made in the critical medical procedure of transplantation. However, persistent challenges such as ischemia-reperfusion injury (IRI) and graft rejection continue to hinder transplant success rates. This comprehensive review explores the potential applications and therapeutic implications of hydrogen in transplantation, shedding light on its role in mitigating IRI, improving graft survival, and modulating immune responses. Through a meticulous analysis encompassing both preclinical and clinical studies, we aim to provide valuable insights into the promising utility of hydrogen as a complementary therapy in transplantation.
Collapse
Affiliation(s)
| | - Hiromichi Naito
- Department of Emergency, Critical Care, and Disaster Medicine, Faculty of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan; (T.O.); (T.N.); (T.H.); (T.H.); (K.A.); (T.A.); (M.H.); (T.Y.); (A.N.)
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Wu C, Zou P, Feng S, Zhu L, Li F, Liu TCY, Duan R, Yang L. Molecular Hydrogen: an Emerging Therapeutic Medical Gas for Brain Disorders. Mol Neurobiol 2023; 60:1749-1765. [PMID: 36567361 DOI: 10.1007/s12035-022-03175-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/14/2022] [Indexed: 12/27/2022]
Abstract
Oxidative stress and neuroinflammation are the main physiopathological changes involved in the initiation and progression of various neurodegenerative disorders or brain injuries. Since the landmark finding reported in 2007 found that hydrogen reduced the levels of peroxynitrite anions and hydroxyl free radicals in ischemic stroke, molecular hydrogen's antioxidative and anti-inflammatory effects have aroused widespread interest. Due to its excellent antioxidant and anti-inflammatory properties, hydrogen therapy via different routes of administration exhibits great therapeutic potential for a wide range of brain disorders, including Alzheimer's disease, neonatal hypoxic-ischemic encephalopathy, depression, anxiety, traumatic brain injury, ischemic stroke, Parkinson's disease, and multiple sclerosis. This paper reviews the routes for hydrogen administration, the effects of hydrogen on the previously mentioned brain disorders, and the primary mechanism underlying hydrogen's neuroprotection. Finally, we discuss hydrogen therapy's remaining issues and challenges in brain disorders. We conclude that understanding the exact molecular target, finding novel routes, and determining the optimal dosage for hydrogen administration is critical for future studies and applications.
Collapse
Affiliation(s)
- Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Peibin Zou
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Shu Feng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Ling Zhu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Fanghui Li
- School of Sports Science, Nanjing Normal University, Nanjing, 210046, China
| | - Timon Cheng-Yi Liu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Rui Duan
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| |
Collapse
|
11
|
Mitroshina EV, Saviuk M, Vedunova MV. Necroptosis in CNS diseases: Focus on astrocytes. Front Aging Neurosci 2023; 14:1016053. [PMID: 36778591 PMCID: PMC9911465 DOI: 10.3389/fnagi.2022.1016053] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/28/2022] [Indexed: 01/28/2023] Open
Abstract
In the last few years, necroptosis, a recently described type of cell death, has been reported to play an important role in the development of various brain pathologies. Necroptosis is a cell death mechanism that has morphological characteristics similar to necrosis but is mediated by fundamentally different molecular pathways. Necroptosis is initiated by signaling through the interaction of RIP1/RIP3/MLKL proteins (receptor-interacting protein kinase 1/receptor-interacting protein kinase 3/mixed lineage kinase domain-like protein). RIPK1 kinase is usually inactive under physiological conditions. It is activated by stimulation of death receptors (TNFR1, TNFR2, TLR3, and 4, Fas-ligand) by external signals. Phosphorylation of RIPK1 results in the formation of its complex with death receptors. Further, complexes with the second member of the RIP3 and MLKL cascade appear, and the necroptosome is formed. There is enough evidence that necroptosis plays an important role in the pathogenesis of brain ischemia and neurodegenerative diseases. In recent years, a point of view that both neurons and glial cells can play a key role in the development of the central nervous system (CNS) pathologies finds more and more confirmation. Astrocytes play complex roles during neurodegeneration and ischemic brain damage initiating both impair and protective processes. However, the cellular and molecular mechanisms that induce pathogenic activity of astrocytes remain veiled. In this review, we consider these processes in terms of the initiation of necroptosis. On the other hand, it is important to remember that like other types of programmed cell death, necroptosis plays an important role for the organism, as it induces a strong immune response and is involved in the control of cancerogenesis. In this review, we provide an overview of the complex role of necroptosis as an important pathogenetic component of neuronal and astrocyte death in neurodegenerative diseases, epileptogenesis, and ischemic brain damage.
Collapse
|
12
|
Pharmacokinetics of hydrogen administered intraperitoneally as hydrogen-rich saline and its effect on ischemic neuronal cell death in the brain in gerbils. PLoS One 2022; 17:e0279410. [PMID: 36574398 PMCID: PMC9794077 DOI: 10.1371/journal.pone.0279410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/06/2022] [Indexed: 12/28/2022] Open
Abstract
Intraperitoneal administration of hydrogen (H2)-containing saline inhibited neuronal cell death in ischemic stroke in a number of animal models, but it is unknown whether H2 is absorbed from the abdominal cavity into the blood and reaches the brain. In this study, we investigated whether intraperitoneal administration of saline containing H2 inhibits neuronal cell death caused by cerebral ischemia and measured the concentration of H2 in the carotid artery and inferior vena cava (IVC). Gerbils were subjected to transient unilateral cerebral ischemia twice, and saline or H2-rich saline was administered intraperitoneally three or seven times every 12 hours. We evaluated the number of apoptotic cells in the hippocampus and cerebral cortex on day 3 and the number of viable neurons in the hippocampus and cerebral cortex on day 7. In addition, a single dose of saline or H2-rich saline was administered intraperitoneally, and blood H2 levels in the carotid artery and IVC were measured. On day 3 of ischemia/reperfusion, the number of neurons undergoing apoptosis in the cortex was significantly lower in the H2-rich saline group than in the saline group, and on day 7, the number of viable neurons in the hippocampus and cerebral cortex was significantly higher in the H2-rich saline group. Intraperitoneal administration of H2-rich saline resulted in large increases in H2 concentration in the IVC ranging from 0.00183 mg/L (0.114%) to 0.00725 mg/L (0.453%). In contrast, carotid H2 concentrations remained in the range of 0.00008 mg/L (0.0049%) to 0.00023 (0.0146%). On average, H2 concentrations in carotid artery were 0.04 times lower than in IVC. These results indicate that intraperitoneal administration of H2-rich saline significantly suppresses neuronal cell death after cerebral ischemia, even though H2 hardly reaches the brain.
Collapse
|
13
|
A potential clinical application of hydrogen-rich saline in patients with traumatic brain injury. J Pediatr Surg 2022; 57:1032. [PMID: 35843727 DOI: 10.1016/j.jpedsurg.2022.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 06/14/2022] [Indexed: 11/27/2022]
|
14
|
Nie Z, Tan L, Niu J, Wang B. The role of regulatory necrosis in traumatic brain injury. Front Mol Neurosci 2022; 15:1005422. [PMID: 36329694 PMCID: PMC9622788 DOI: 10.3389/fnmol.2022.1005422] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/27/2022] [Indexed: 11/18/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability in the population worldwide, of which key injury mechanism involving the death of nerve cells. Many recent studies have shown that regulatory necrosis is involved in the pathological process of TBI which includes necroptosis, pyroptosis, ferroptosis, parthanatos, and Cyclophilin D (CypD) mediated necrosis. Therefore, targeting the signaling pathways involved in regulatory necrosis may be an effective strategy to reduce the secondary injury after TBI. Meanwhile, drugs or genes are used as interference factors in various types of regulatory necrosis, so as to explore the potential treatment methods for the secondary injury after TBI. This review summarizes the current progress on regulatory necrosis in TBI.
Collapse
|
15
|
Lu W, Zhu Z, Shi D, Li X, Luo J, Liao X. Cerebrolysin alleviates early brain injury after traumatic brain injury by inhibiting neuroinflammation and apoptosis via TLR signaling pathway. Acta Cir Bras 2022; 37:e370605. [PMID: 36074398 PMCID: PMC9448247 DOI: 10.1590/acb370605] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 05/03/2022] [Indexed: 11/22/2022] Open
Abstract
Purpose: Traumatic brain injury (TBI) is a major cause of death and disability. Cerebrolysin (CBL) has been reported to be anti-inflammatory by reducing reactive oxygen species (ROS) production. However, the neuroprotection of CBL in TBI and the potential mechanism are unclear. We aimed to investigate the neuroprotection and mechanisms of CBL in TBI. Methods: The TBI model was established in strict accordance with the Feeney weight-drop model of focal injury. The neurological score, brain water content, neuroinflammatory cytokine levels, and neuronal damage were evaluated. The involvement of the early brain injury modulatory pathway was also investigated. Results: Following TBI, the results showed that CBL administration increased neurological scores and decreased brain edema by alleviating blood‑brain barrier (BBB) permeability, upregulating tight junction protein (ZO‑1) levels, and decreasing the levels of the inflammatory cytokines tumor necrosis factor‑α (TNF‑α), interleukin‑1β (IL‑1β), IL‑6, and NF‑κB. The TUNEL assay showed that CBL decreased hippocampal neuronal apoptosis after TBI and decreased the protein expression levels of caspase‑3 and Bax, increasing the levels of Bcl‑2. The levels of Toll‑like receptor 2 (TLR2) and TLR4 were significantly decreased after CBL treatment. In TBI patients, CBL can also decrease TNF‑α, IL‑1β, IL‑6, and NF‑κB levels. This result indicates that CBL‑mediated inhibition of neuroinflammation and apoptosis ameliorated neuronal death after TBI. The neuroprotective capacity of CBL is partly dependent on the TLR signaling pathway. Conclusions: Taken together, the results of this study indicate that CBL can improve neurological outcomes and reduce neuronal death against neuroinflammation and apoptosis via the TLR signaling pathway in mice.
Collapse
Affiliation(s)
- Weihong Lu
- BS. 904th Hospital of Joint Logistic Support Force of PLA - Department of Anesthesiology - Wuxi, China
| | - Zhonghua Zhu
- BS. 904th Hospital of Joint Logistic Support Force of PLA - Department of Anesthesiology - Wuxi, China
| | - Dongliang Shi
- MD. Anhui Medical University - Wuxi Clinical College - 904th Hospital of Joint Logistic Support Force of PLA - Department of Neurosurgery - Wuxi, China
| | - Xiaoyu Li
- BS. 904th Hospital of Joint Logistic Support Force of PLA - Department of Anesthesiology - Wuxi, China
| | - Jingzhi Luo
- BS. 904th Hospital of Joint Logistic Support Force of PLA - Department of Anesthesiology - Wuxi, China
| | - Xingzhi Liao
- BS. 904th Hospital of Joint Logistic Support Force of PLA - Department of Anesthesiology - Wuxi, China
| |
Collapse
|
16
|
Wu X, Jiao W, Chen J, Tao Y, Zhang J, Wang Y. Ulinastatin alleviates early brain injury after intracerebral hemorrhage by inhibiting oxidative stress and neuroinflammation via ROS/MAPK/Nrf2 signaling pathway. Acta Cir Bras 2022; 37:e370606. [PMID: 36074399 PMCID: PMC9448248 DOI: 10.1590/acb370606] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 05/11/2022] [Indexed: 11/26/2022] Open
Abstract
Purpose: Spontaneous intracerebral hemorrhage (ICH) is still a major public health problem, with high mortality and disability. Ulinastatin (UTI) was purified from human urine and has been reported to be anti-inflammatory, organ protective, and antioxidative stress. However, the neuroprotection of UTI in ICH has not been confirmed, and the potential mechanism is unclear. In the present study, we aimed to investigate the neuroprotection and potential molecular mechanisms of UTI in ICH-induced early brain injury in a C57BL/6 mouse model. Methods: The neurological score, brain water content, neuroinflammatory cytokine levels, oxidative stress levels, and neuronal damage were evaluated. Results: UTI treatment markedly increased the neurological score, alleviated brain edema, decreased the levels of the inflammatory cytokines tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-6, and NF-κB, decreased the levels of reactive oxygen species (ROS) and malondialdehyde (MDA), and upregulated the levels of glutathione (GSH), superoxide dismutase (SOD), and Nrf2. This finding indicated that UTI-mediated inhibition of neuroinflammation and oxidative stress alleviated neuronal damage after ICH. The neuroprotective capacity of UTI is partly dependent on the ROS/MAPK/Nrf2 signaling pathway. Conclusions: UTI improves neurological outcomes in mice and reduces neuronal death by protecting against neural neuroinflammation and oxidative stress.
Collapse
Affiliation(s)
- Xi Wu
- BS. 904th Hospital of Joint Logistic Support Force of PLA - Anhui Medical University - Wuxi Clinical College - Department of Neurosurgery - Wuxi, China
| | - Wei Jiao
- MD. 904th Hospital of Joint Logistic Support Force of PLA - Anhui Medical University - Wuxi Clinical College - Department of Neurosurgery - Wuxi, China
| | - Junhui Chen
- BS. 904th Hospital of Joint Logistic Support Force of PLA - Anhui Medical University - Wuxi Clinical College - Department of Neurosurgery - Wuxi, China
| | - Yunna Tao
- BS. 904th Hospital of Joint Logistic Support Force of PLA - Anhui Medical University - Wuxi Clinical College - Department of Neurosurgery - Wuxi, China
| | - Jing Zhang
- BS. 904th Hospital of Joint Logistic Support Force of PLA - Anhui Medical University - Wuxi Clinical College - Department of Neurosurgery - Wuxi, China
| | - Yuhai Wang
- PhD. 904th Hospital of Joint Logistic Support Force of PLA - Anhui Medical University - Wuxi Clinical College - Department of Neurosurgery - Wuxi, China
| |
Collapse
|
17
|
Neuroprotective and Anti-inflammatory Effects of Pioglitazone on Traumatic Brain Injury. Mediators Inflamm 2022; 2022:9860855. [PMID: 35757108 PMCID: PMC9232315 DOI: 10.1155/2022/9860855] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/06/2022] [Accepted: 05/24/2022] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury (TBI) is still a major cause of concern for public health, and out of all the trauma-related injuries, it makes the highest contribution to death and disability worldwide. Patients of TBI continue to suffer from brain injury through an intricate flow of primary and secondary injury events. However, when treatment is provided in a timely manner, there is a significant window of opportunity to avoid a few of the serious effects. Pioglitazone (PG), which has a neuroprotective impact and can decrease inflammation after TBI, activates peroxisome proliferator-activated receptor-gamma (PPARγ). The objective of the study is to examine the existing literature to assess the neuroprotective and anti-inflammatory impact of PG in TBI. It also discusses the part played by microglia and cytokines in TBI. According to the findings of this study, PG has the ability to enhance neurobehavior, decrease brain edema and neuronal injury following TBI. To achieve the protective impact of PG the following was required: (1) stimulating PPARγ; (2) decreasing oxidative stress; (3) decreasing nuclear factor kappa B (NF-κB), interleukin 6 (IL-6), interleukin-1β (IL-1β), cyclooxygenase-2 (COX-2), and C-C motif chemokine ligand 20 (CCL20) expression; (4) limiting the increase in the number of activated microglia; and (5) reducing mitochondrial dysfunction. The findings indicate that when PIG is used clinically, it may serve as a neuroprotective anti-inflammatory approach in TBI.
Collapse
|