1
|
He Q, Lan X, Ding M, Zhang N. Long-term consumption of hydrogen-rich water provides hepatoprotection by improving mitochondrial biology and quality control in chronically stressed mice. PLoS One 2025; 20:e0317080. [PMID: 39951412 PMCID: PMC11828380 DOI: 10.1371/journal.pone.0317080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 12/20/2024] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Chronic stress has emerged as a prevalent facet of contemporary existence, significantly jeopardizing overall bodily health. The liver, a pivotal organ responsible for metabolic equilibrium, is particularly vulnerable to its adverse effects. This study delves into the hepatoprotective properties of extended consumption of HRW in mice subjected to chronic stress. METHODS Mice subjected to chronic stress via CUMS and HRW administration for seven months underwent liver pathological examination. Key liver function indicators (AST, ALT), oxidative stress markers (SOD, CAT, GSH), and markers related to lipid peroxidation and ferroptosis (MDA, Fe) were measured using standard kits. ELISA determined corticosterone and 4-HNE levels. Immunofluorescence evaluated ROS, Nrf2, and apoptosis in liver tissues. Western blotting analyzed markers for ferroptosis (GPX4, SLC7A11, HO-1, Nrf2), apoptosis (Bax, Bcl-2, Cytc, Caspase-3, Caspase-8), mitochondrial biogenesis (Nrf1, PGC-1α, Tfam), and quality control (Drp1, Fis1, Mfn1, Mfn2, OPA1, PINK1, Parkin, LC3 I/II). RESULTS The findings indicate a noteworthy improvement in liver health among mice exposed to HRW, as evidenced by histological analysis. Furthermore, the consumption of HRW exhibited hepatoprotection, as evidenced by the normalization of AST and ALT levels. Mechanistically, our results indicate that HRW elevates the levels of SOD, CAT, and GSH, while effectively clearing ROS within mitochondria. It was observed led to a regulation in the expression of mitochondrial quality control proteins, consequently improving mitochondrial biogenesis (Nrf1, PGC-1α, Tfam), and increasing ATP production. Furthermore, HRW decreased Cytc, Bax, Caspase-3, and Caspase-8 levels, and increasing the expression of Bcl-2. Additionally, HRW reduced MDA and 4-HNE levels, alleviating ferroptosis through the Nrf2/HO-1 pathway, and upregulating the expression of GPX4 and SLC7A11. By mitigating hepatocyte death through the aforementioned mechanisms, HRW fulfills its crucial role in safeguarding liver health. CONCLUSIONS This study reveals that long-term hydrogen-rich water (HRW) consumption provides significant hepatoprotection in mice under chronic stress. HRW normalizes liver enzyme levels, enhances antioxidant capacity, and reduces lipid peroxidation and ferroptosis. It improves mitochondrial biogenesis, function, and ATP production, and attenuates apoptosis by modulating related proteins. Behavioral tests show HRW alleviates stress-induced anxiety and enhances exploratory behavior. These findings suggest HRW is a promising non-invasive intervention for preventing and treating stress-related liver disorders by targeting oxidative stress and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Qi He
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Xiang Lan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Mengyuan Ding
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Na Zhang
- The Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, China
| |
Collapse
|
2
|
Martínez-Martel I, Pol O. A Novel Therapy for Cisplatin-Induced Allodynia and Dysfunctional and Emotional Impairments in Male and Female Mice. Antioxidants (Basel) 2023; 12:2063. [PMID: 38136183 PMCID: PMC10741113 DOI: 10.3390/antiox12122063] [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: 10/26/2023] [Revised: 11/23/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Patients undergoing chemotherapy with cisplatin (CIS) develop neuropathy in addition to other symptoms such as, anxiety, depression, muscle wasting and body weight loss. This symptomatology greatly weakens patients and may even lead to adjournment of chemotherapy. The protecting actions of molecular hydrogen in many neurological illnesses have been described, but its effect on the functional and emotional deficiencies caused by CIS has not been assessed. In C57BL/6J male and female mice injected with CIS, we examined the impact of the prophylactic treatment with hydrogen-rich water (HRW) on: (i) the tactile and cold allodynia, (ii) the deficits of grip strength and weight loss, (iii) the anxiodepressive-like behaviors and (iv) the inflammatory and oxidative reactions incited by CIS in the dorsal root ganglia (DRG) and prefrontal cortex (PFC). The results demonstrate that the mechanical allodynia and the anxiodepressive-like comportment provoked by CIS were similarly manifested in both sexes, whereas the cold allodynia, grip strength deficits and body weight loss produced by this chemotherapeutic agent were greater in female mice. Nonetheless, the prophylactic treatment with HRW prevented the allodynia and the functional and emotional impairments resulting from CIS in both sexes. This treatment also inhibited the inflammatory and oxidative responses activated by CIS in the DRG and PFC in both sexes, which might explain the therapeutic actions of HRW in male and female mice. In conclusion, this study revealed the plausible use of HRW as a new therapy for the allodynia and physical and mental impairments linked with CIS and its possible mechanism of action.
Collapse
Affiliation(s)
- Ignacio Martínez-Martel
- Grup de Neurofarmacologia Molecular, Institut de Recerca Sant Pau, Sant Quintí 77-79, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Olga Pol
- Grup de Neurofarmacologia Molecular, Institut de Recerca Sant Pau, Sant Quintí 77-79, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| |
Collapse
|
3
|
Chen Y, Yan P, Wei S, Zhu Y, Lai J, Zhou Q. Ketamine metabolite alleviates morphine withdrawal-induced anxiety via modulating nucleus accumbens parvalbumin neurons in male mice. Neurobiol Dis 2023; 186:106279. [PMID: 37661023 DOI: 10.1016/j.nbd.2023.106279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/20/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023] Open
Abstract
Opioid withdrawal generates extremely unpleasant physical symptoms and negative affective states. A rapid relief of opioid withdrawal-induced anxiety has obvious clinical relevance but has been rarely reported. We have shown that injection of ketamine metabolite (2R,6R)-hydroxynorketamine (HNK) leads to a rapid alleviation of anxiety-like behaviors in male mice undergoing chronic morphine withdrawal. Here we investigated the contribution of nucleus accumbens shell (sNAc) parvalbumin (PV)-neurons to this process. Chronic morphine withdrawal was associated with higher intrinsic excitability of sNAc PV-neurons via reduced voltage-dependent potassium currents. Chemogenetic inhibition of sNAc PV-neurons reversed the enhanced excitability of PV-neurons and anxiety-like behaviors in these morphine withdrawal male mice, while activation of sNAc PV-neurons induced anxiety-like behaviors in naive male mice. (2R,6R)-HNK reversed the altered potassium currents and intrinsic excitability of sNAc PV-neurons. Our findings demonstrate an important contribution of sNAc PV-neurons to modulating morphine withdrawal-induced anxiety-like behaviors and rapid relief of anxiety-like behaviors by (2R,6R)-HNK, this newly identified target may have therapeutic potentials in treating opioid addiction and anxiety disorders.
Collapse
Affiliation(s)
- Yuanyuan Chen
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, China; School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Peng Yan
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Shuguang Wei
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Yongsheng Zhu
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Jianghua Lai
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, China.
| | - Qiang Zhou
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China.
| |
Collapse
|
4
|
Coral-Pérez S, Martínez-Martel I, Martínez-Serrat M, Batallé G, Bai X, Leite-Panissi CRA, Pol O. Treatment with Hydrogen-Rich Water Improves the Nociceptive and Anxio-Depressive-like Behaviors Associated with Chronic Inflammatory Pain in Mice. Antioxidants (Basel) 2022; 11:2153. [PMID: 36358525 PMCID: PMC9686765 DOI: 10.3390/antiox11112153] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/24/2022] [Accepted: 10/28/2022] [Indexed: 09/25/2023] Open
Abstract
Chronic inflammatory pain is manifested in many diseases. The potential use of molecular hydrogen (H2) as a new therapy for neurological disorders has been demonstrated. Recent studies prove its analgesic properties in animals with neuropathic pain, but the possible antinociceptive, antidepressant, and/or anxiolytic actions of H2 during persistent inflammatory pain have not been investigated. Therefore, using male mice with chronic inflammatory pain incited by the subplantar injection of complete Freud's adjuvant (CFA), we assessed the actions of hydrogen-rich water (HRW) systemically administered on: (1) the nociceptive responses and affective disorders associated and (2) the oxidative (4-hydroxy-2-nonenal; 4-HNE), inflammatory (phosphorylated-NF-kB inhibitor alpha; p-IKBα), and apoptotic (Bcl-2-like protein 4; BAX) changes provoked by CFA in the paws and amygdala. The role of the antioxidant system in the analgesia induced by HRW systemically and locally administered was also determined. Our results revealed that the intraperitoneal administration of HRW, besides reducing inflammatory pain, also inhibited the depressive- and anxiolytic-like behaviors associated and the over expression of 4-HNE, p-IKBα, and BAX in paws and amygdala. The contribution of the nuclear factor erythroid 2-related factor 2/heme oxygenase 1 and NAD(P)H: quinone oxidoreductase 1 pathway in the analgesic activities of HRW, systemically or locally administered, was also shown. These data revealed the analgesic, antidepressant, and anxiolytic actions of HRW. The protective, anti-inflammatory, and antioxidant qualities of this treatment during inflammatory pain were also demonstrated. Therefore, this study proposes the usage of HRW as a potential therapy for chronic inflammatory pain and linked comorbidities.
Collapse
Affiliation(s)
- Santiago Coral-Pérez
- Grup de Neurofarmacologia Molecular, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Ignacio Martínez-Martel
- Grup de Neurofarmacologia Molecular, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Maria Martínez-Serrat
- Grup de Neurofarmacologia Molecular, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Gerard Batallé
- Grup de Neurofarmacologia Molecular, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Xue Bai
- Grup de Neurofarmacologia Molecular, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Christie R. A. Leite-Panissi
- Department of Psychology, Faculty of Philosophy Science and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
| | - Olga Pol
- Grup de Neurofarmacologia Molecular, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| |
Collapse
|
5
|
Martínez-Serrat M, Martínez-Martel I, Coral-Pérez S, Bai X, Batallé G, Pol O. Hydrogen-Rich Water as a Novel Therapeutic Strategy for the Affective Disorders Linked with Chronic Neuropathic Pain in Mice. Antioxidants (Basel) 2022; 11:antiox11091826. [PMID: 36139900 PMCID: PMC9495356 DOI: 10.3390/antiox11091826] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022] Open
Abstract
Neuropathic pain manifested with allodynia and hyperalgesia usually becomes a chronic condition accompanied with mood disorders. Clinical therapies for neuropathic pain are still unsatisfactory with notable side effects. Recent studies have reported the protective role of molecular hydrogen (H2) in different diseases including neurological disorders, such as Alzheimer's as well as its antidepressant activities in animals with chronic stress. This study explored the effects of treatment with hydrogen-rich water (HRW) in male mice with neuropathic pain induced by the chronic constriction of the sciatic nerve (CCI) and the accompanying affective deficits. The likely pathways implied in the HRW analgesic activity, as well as the interaction between heme oxygenase 1 (HO-1) enzyme and H2 during neuropathic pain were also studied. The results showed: (i) the inhibitory effects of the repetitive treatment with HRW on the allodynia and hyperalgesia provoked by CCI; (ii) the anxiolytic and antidepressant actions of HRW in animals with neuropathic pain; (iii) the contribution of the antioxidant enzymes (HO-1 and NAD(P)H: quinone oxidoreductase 1) and the ATP sensitive potassium channels in the painkiller activities of HRW during neuropathic pain; (iv) a positive interaction between the HO-1 and H2 systems in inhibiting the CCI-induced neuropathy; and (v) the antioxidant, antinociceptive, anti-inflammatory and/or antiapoptotic features of HRW treatment in the dorsal root ganglia and/or amygdala of sciatic nerve-injured mice. This study demonstrates new protective actions of H2 and suggests that treatment with HRW might be an interesting therapeutic strategy for chronic neuropathic pain and its associated mood disorders.
Collapse
Affiliation(s)
- Maria Martínez-Serrat
- Grup de Neurofarmacologia Molecular, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Ignacio Martínez-Martel
- Grup de Neurofarmacologia Molecular, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Santiago Coral-Pérez
- Grup de Neurofarmacologia Molecular, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Xue Bai
- Grup de Neurofarmacologia Molecular, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Gerard Batallé
- Grup de Neurofarmacologia Molecular, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Olga Pol
- Grup de Neurofarmacologia Molecular, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Correspondence: ; Tel.: +34-619-757-054
| |
Collapse
|
6
|
Neuwirth LS, Verrengia MT, Harikinish-Murrary ZI, Orens JE, Lopez OE. Under or Absent Reporting of Light Stimuli in Testing of Anxiety-Like Behaviors in Rodents: The Need for Standardization. Front Mol Neurosci 2022; 15:912146. [PMID: 36061362 PMCID: PMC9428565 DOI: 10.3389/fnmol.2022.912146] [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: 04/04/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
Behavioral neuroscience tests such as the Light/Dark Test, the Open Field Test, the Elevated Plus Maze Test, and the Three Chamber Social Interaction Test have become both essential and widely used behavioral tests for transgenic and pre-clinical models for drug screening and testing. However, as fast as the field has evolved and the contemporaneous involvement of technology, little assessment of the literature has been done to ensure that these behavioral neuroscience tests that are crucial to pre-clinical testing have well-controlled ethological motivation by the use of lighting (i.e., Lux). In the present review paper, N = 420 manuscripts were examined from 2015 to 2019 as a sample set (i.e., n = ~20–22 publications per year) and it was found that only a meager n = 50 publications (i.e., 11.9% of the publications sampled) met the criteria for proper anxiogenic and anxiolytic Lux reported. These findings illustrate a serious concern that behavioral neuroscience papers are not being vetted properly at the journal review level and are being released into the literature and public domain making it difficult to assess the quality of the science being reported. This creates a real need for standardizing the use of Lux in all publications on behavioral neuroscience techniques within the field to ensure that contributions are meaningful, avoid unnecessary duplication, and ultimately would serve to create a more efficient process within the pre-clinical screening/testing for drugs that serve as anxiolytic compounds that would prove more useful than what prior decades of work have produced. It is suggested that improving the standardization of the use and reporting of Lux in behavioral neuroscience tests and the standardization of peer-review processes overseeing the proper documentation of these methodological approaches in manuscripts could serve to advance pre-clinical testing for effective anxiolytic drugs. This report serves to highlight this concern and proposes strategies to proactively remedy them as the field moves forward for decades to come.
Collapse
Affiliation(s)
- Lorenz S. Neuwirth
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
- *Correspondence: Lorenz S. Neuwirth
| | - Michael T. Verrengia
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
| | - Zachary I. Harikinish-Murrary
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
| | - Jessica E. Orens
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
| | - Oscar E. Lopez
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
| |
Collapse
|
7
|
Shi Z, Pan S, Wang L, Li S. Oleanolic Acid Attenuates Morphine Withdrawal Symptoms in Rodents: Association with Regulation of Dopamine Function. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:3685-3696. [PMID: 34465980 PMCID: PMC8402955 DOI: 10.2147/dddt.s326583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/09/2021] [Indexed: 01/13/2023]
Abstract
Introduction Oleanolic acid (OA) has been shown to be useful for the treatment of mental disorders. Methods In this study, we investigated the effects of OA in animal models of spontaneous withdrawal and naloxone-precipitated withdrawal and evaluated the effects of OA on the acquisition, extinction, and reinstatement of morphine-induced conditioned place preference (CPP). Results OA significantly improved symptoms of withdrawal, and significantly reduced the acquisition and reinstatement of morphine-induced conditioned place preference. Moreover, OA significantly reduced the serum content of 5-hydroxy tryptamine (5-HT) and dopamine (DA) in a dose-dependent manner, and reduced norepinephrine (NE) and 5-HT content in the frontal cortex (PFC), while significantly increasing endorphin content in rats. OA also significantly reduced serum DA content in mice. Conclusion These results indicate that OA can improve the withdrawal symptoms of rats and mice by regulating the DA system and suggest that OA may be useful in treatment of morphine addiction.
Collapse
Affiliation(s)
- Zhiqi Shi
- School of Pharmacy, Changzhou Institute of Industry and Technology, Changzhou, Jiangsu, People's Republic of China.,Longsha Medical Research Institute, Wuxi Hospital of Traditional Chinese Medicine, Wuxi, Jiangsu, People's Republic of China
| | - Shugang Pan
- School of Pharmacy, Changzhou Institute of Technology, Changzhou, 213022, People's Republic of China.,Key Laboratory for Soft Chemistry and Functional Materials of Ministry Education, Nanjing University of Science and Technology, Nanjing, People's Republic of China
| | - Luolin Wang
- Department of Pharmacy, Guangdong Provincial Institute of Traditional Chinese Medicine, Guangzhou, People's Republic of China
| | - Sha Li
- Longsha Medical Research Institute, Wuxi Hospital of Traditional Chinese Medicine, Wuxi, Jiangsu, People's Republic of China
| |
Collapse
|
8
|
Satoh Y. The Potential of Hydrogen for Improving Mental Disorders. Curr Pharm Des 2021; 27:695-702. [PMID: 33185151 DOI: 10.2174/1381612826666201113095938] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/20/2020] [Indexed: 11/22/2022]
Abstract
In 2007, Ohsawa and colleagues reported that molecular hydrogen (H2) gas significantly reduced the infarct volume size in a rat model of cerebral infarction, which was, at least, partially due to scavenging hydroxyl radicals. Since then, multiple studies have shown that H2 has not only anti-oxidative but also anti-inflammatory and anti-apoptotic properties, which has ignited interest in the clinical use of H2 in diverse diseases. A growing body of studies has indicated that H2 affects both mental and physical conditions. Mental disorders are characterized by disordered mood, thoughts, and behaviors that affect the ability to function in daily life. However, there is no sure way to prevent mental disorders. Although antidepressant and antianxiety drugs relieve symptoms of depression and anxiety, they have efficacy limitations and are accompanied by a wide range of side effects. While mental disorders are generally thought to be caused by a variety of genetic and/or environmental factors, recent progress has shown that these disorders are strongly associated with increased oxidative and inflammatory stress. Thus, H2 has received much attention as a novel therapy for the prevention and treatment of mental disorders. This review summarizes the recent progress in the use of H2 for the treatment of mental disorders and other related diseases. We also discuss the potential mechanisms of the biomedical effects of H2 and conclude that H2 could offer relief to people suffering from mental disorders.
Collapse
Affiliation(s)
- Yasushi Satoh
- Department of Biochemistry, National Defense Medical College, Tokorozawa, Saitama, Japan
| |
Collapse
|
9
|
Co-administration of nalbuphine attenuates the morphine-induced anxiety and dopaminergic alterations in morphine-withdrawn rats. Psychopharmacology (Berl) 2021; 238:1193-1211. [PMID: 33655408 DOI: 10.1007/s00213-021-05765-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 01/12/2021] [Indexed: 10/22/2022]
Abstract
INTRODUCTION The classical effects of exogenous opioids, such as morphine, are predominantly mediated through μ-opioid receptors. The chronic use of morphine induces anxiety-like behavior causing functional changes in the mesolimbic dopaminergic system. The mixed μ/κ-agonist, nalbuphine, used either as an analgesic or as an adjuvant with morphine, produces different and opposite effects. However, whether nalbuphine can be used to antagonize morphine-induced anxiety and dopaminergic alterations is not fully known. OBJECTIVE This study aimed to compare acute and chronic effects of nalbuphine on morphine-induced anxiety and dopaminergic alterations in rats. METHODS Male adult Wistar albino rats were made opioid-dependent by administering increasing doses of morphine (5-25 mg/kg; i.p.; b.i.d.). Withdrawal was induced by naloxone (1 mg/kg, i.p.), 4 h after the last morphine injection. Anxiety-like behavior was measured using Activity Monitor (Coulbourn Instruments, Inc. USA). Thereafter, the animals were sacrificed and the brain dissected out and the level of cAMP and the transcriptional and translational expression of TH was measured. Nalbuphine was co-administered with morphine, acutely and chronically, at various doses (0.1, 0.3, 1.0, 3.0 mg/kg, i.p.). RESULTS Morphine-dependent rats showed a significant higher anxiety and cAMP levels and a significant decrease in the expression of TH. Co-administration of chronic doses of nalbuphine attenuates the higher anxiety, cAMP levels, and upregulates the TH expressions; however, the acute nalbuphine treatment does not attenuate the morphine-induced side effects. CONCLUSION Therefore, nalbuphine might have an important role in attenuating the anxiety and the effects of the dopaminergic pathway and may have potential in the treatment of opioid addiction.
Collapse
|
10
|
Chen W, Zhang HT, Qin SC. Neuroprotective Effects of Molecular Hydrogen: A Critical Review. Neurosci Bull 2021; 37:389-404. [PMID: 33078374 PMCID: PMC7954968 DOI: 10.1007/s12264-020-00597-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 05/31/2020] [Indexed: 12/15/2022] Open
Abstract
Molecular hydrogen (H2) is a physiologically inert gas. However, during the last 10 years, increasing evidence has revealed its biological functions under pathological conditions. More specifically, H2 has protective effects against a variety of diseases, particularly nervous system disorders, which include ischemia/reperfusion injury, traumatic injury, subarachnoid hemorrhage, neuropathic pain, neurodegenerative diseases, cognitive dysfunction induced by surgery and anesthesia, anxiety, and depression. In addition, H2 plays protective roles mainly through anti-oxidation, anti-inflammation, anti-apoptosis, the regulation of autophagy, and preservation of mitochondrial function and the blood-brain barrier. Further, H2 is easy to use and has neuroprotective effects with no major side-effects, indicating that H2 administration is a potential therapeutic strategy in clinical settings. Here we summarize the H2 donors and their pharmacokinetics. Meanwhile, we review the effectiveness and safety of H2 in the treatment of various nervous system diseases based on preclinical and clinical studies, leading to the conclusion that H2 can be a simple and effective clinical therapy for CNS diseases such as ischemia-reperfusion brain injury, Parkinson's disease, and diseases characterized by cognitive dysfunction. The potential mechanisms involved in the neuroprotective effect of H2 are also analyzed.
Collapse
Affiliation(s)
- Wei Chen
- Taishan Institute for Hydrogen Biomedicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, 271000, China
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, 271000, China
| | - Han-Ting Zhang
- Departments of Neuroscience and Behavioral Medicine and Psychiatry, Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, WV, 26506, USA.
| | - Shu-Cun Qin
- Taishan Institute for Hydrogen Biomedicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, 271000, China.
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, 271000, China.
| |
Collapse
|
11
|
Fernández-Teruel A, Tobeña A. Revisiting the role of anxiety in the initial acquisition of two-way active avoidance: pharmacological, behavioural and neuroanatomical convergence. Neurosci Biobehav Rev 2020; 118:739-758. [PMID: 32916193 DOI: 10.1016/j.neubiorev.2020.08.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 07/10/2020] [Accepted: 08/15/2020] [Indexed: 12/19/2022]
Abstract
Two-way active avoidance (TWAA) acquisition constitutes a particular case of approach -avoidance conflict for laboratory rodents. The present article reviews behavioural, psychopharmacological and neuroanatomical evidence accumulated along more than fifty years that provides strong support to the contention that anxiety is critical in the transition from CS (conditioned stimulus)-induced freezing to escape/avoidance responses during the initial stages of TWAA acquisition. Thus, anxiolytic drugs of different types accelerate avoidance acquisition, anxiogenic drugs impair it, and avoidance during these initial acquisition stages is negatively associated with other typical measures of anxiety. In addition behavioural and developmental treatments that reduce or increase anxiety/stress respectively facilitate or impair TWAA acquisition. Finally, evidence for the regulation of TWAA acquisition by septo-hippocampal and amygdala-related mechanisms is discussed. Collectively, the reviewed evidence gives support to the initial acquisition of TWAA as a paradigm with considerable predictive and (in particular) construct validity as an approach-avoidance conflict-based rodent anxiety model.
Collapse
Affiliation(s)
- Alberto Fernández-Teruel
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, School of Medicine, Autonomous University of Barcelona, 08193-Bellaterra, Barcelona, Spain.
| | - Adolf Tobeña
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, School of Medicine, Autonomous University of Barcelona, 08193-Bellaterra, Barcelona, Spain
| |
Collapse
|
12
|
Wen D, Hui R, Liu Y, Luo Y, Wang J, Shen X, Xie B, Yu F, Cong B, Ma C. Molecular hydrogen attenuates methamphetamine-induced behavioral sensitization and activation of ERK-ΔFosB signaling in the mouse nucleus accumbens. Prog Neuropsychopharmacol Biol Psychiatry 2020; 97:109781. [PMID: 31629777 DOI: 10.1016/j.pnpbp.2019.109781] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/30/2019] [Accepted: 10/06/2019] [Indexed: 12/20/2022]
Abstract
Methamphetamine (METH) is one of the most prevalently used illegal psychostimulants in many countries. Continuous exposure to METH leads to behavioral sensitization in animals, which can be used as a behavioral model with many mechanisms in common with relapse in humans. Molecular hydrogen has recently gained attention for its potential as a novel healthcare product with preventive and therapeutic applicability to a wide range of pathological conditions. However, it remains unclear whether and, if so, how hydrogen regulates METH-induced behavioral abnormalities. In the present study, we investigated the roles of molecular hydrogen on the acquisition and transfer of METH-induced behavioral sensitization and the accompanying changes in ERK phosphorylation and ΔFosB activation in the nucleus accumbens (NAc) of mice. To this end, male C57BL/6 mice received METH (0.1, 0.5 and 1.0 mg/kg, i.p.) injections for 7 days followed by a METH challenge (0.1, 0.5 and 1.0 mg/kg, i.p.) after a 7-day transfer period. Molecular hydrogen, delivered through a hydrogen-rich saline (HRS) injection (10 mL/kg, i.p., 3-h interval), was administered during the acquisition and transfer periods. We found that HRS administration was able to inhibit the acquisition and transfer of 0.1 and 0.5 mg/kg METH-induced behavioral sensitization to a certain extent, thereby attenuating the expression of behavioral sensitization. The HRS injections alone did not induce any obvious changes in locomotor activity in mice. Intriguingly, the increases in pERK and ΔFosB in the NAc, which accompanied the METH-induced behavioral sensitization, were also attenuated by the HRS treatments. Due to the anti-oxidative function of molecular hydrogen, the HRS injections reduced METH-induced reactive oxygen species and malondialdehyde generation in the NAc. These results suggest that molecular hydrogen serves as an anti-oxidative agent with potentially therapeutic applicability to the treatment of METH addicts.
Collapse
Affiliation(s)
- Di Wen
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Province, Shijiazhuang 050017, PR China
| | - Rongji Hui
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Province, Shijiazhuang 050017, PR China
| | - Yi Liu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Province, Shijiazhuang 050017, PR China
| | - Yixiao Luo
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410081,China
| | - Jian Wang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Province, Shijiazhuang 050017, PR China
| | - Xi Shen
- College of Public Health, Hebei Medical University, Hebei Province, Shijiazhuang 050017, PR China
| | - Bing Xie
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Province, Shijiazhuang 050017, PR China
| | - Feng Yu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Province, Shijiazhuang 050017, PR China
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Province, Shijiazhuang 050017, PR China.
| | - Chunling Ma
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Province, Shijiazhuang 050017, PR China.
| |
Collapse
|
13
|
Martins CC, Rosa SG, Recchi AMS, Nogueira CW, Zeni G. m-Trifluoromethyl-diphenyl diselenide (m-CF 3-PhSe) 2 modulates the hippocampal neurotoxic adaptations and abolishes a depressive-like phenotype in a short-term morphine withdrawal in mice. Prog Neuropsychopharmacol Biol Psychiatry 2020; 98:109803. [PMID: 31689445 DOI: 10.1016/j.pnpbp.2019.109803] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/17/2019] [Accepted: 10/31/2019] [Indexed: 12/21/2022]
Abstract
The opioid withdrawal syndrome is defined as a complex phenomenon involving multiple cellular adaptations, which leads to the emergence of aversive physical and affective signs. The m-trifluoromethyl-diphenyl diselenide (m-CF3-PhSe)2 elicits an antidepressant-like effect by modulating the opioid system in different animal models of mood disorders. Notably, repeated exposure to (m-CF3-PhSe)2 developed neither tolerance nor withdrawal signs in mice. The aim of the present study was to investigate whether (m-CF3-PhSe)2 attenuates the physical signs and the depressive-like phenotype during morphine withdrawal through its neuroprotective effects on oxidative stress, the NMDA receptor and the proBDNF/mBDNF signaling in the hippocampus of mice. Adult Swiss mice received saline solution or escalating doses (20-100 mg/kg, sc) of morphine for six days. For the next three days, the animals were treated with canola oil, (m-CF3-PhSe)2 (5 and 10 mg/kg, ig) or methadone (5 mg/kg, sc) whereas morphine injections were discontinued. On day 9, physical withdrawal signs and depressive-like behavior were assessed 30 min after the last administration of (m-CF3-PhSe)2. Although short-term treatment with (m-CF3-PhSe)2 at both doses suppressed the aversive physical and affective signs in morphine withdrawn-mice, the highest dose of (m-CF3-PhSe)2 per se increased the teeth chattering manifestation. The intrinsic antioxidant property of (m-CF3-PhSe)2 modulated oxidative stress, it also restored the NMDA receptor levels in the hippocampus of morphine withdrawn-mice. Besides, (m-CF3-PhSe)2 downregulated the proBDNF/p-75NTR/JNK pro-apoptotic pathway without affecting the mBDNF/TrkB/ERK/CREB pro-survival signaling in the hippocampus of morphine withdrawn-mice. The results show that (m-CF3-PhSe)2 treatment modulated the hippocampal neurotoxic adaptations and abolished the depressive-like phenotype following morphine withdrawal in mice.
Collapse
Affiliation(s)
- Carolina C Martins
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Suzan G Rosa
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Ana M S Recchi
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Cristina W Nogueira
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Gilson Zeni
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil.
| |
Collapse
|
14
|
Abstract
This paper is the fortieth consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2017 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, CUNY, 65-30 Kissena Blvd., Flushing, NY, 11367, United States.
| |
Collapse
|
15
|
Lu S, Liao L, Zhang B, Yan W, Chen L, Yan H, Guo L, Lu S, Xiong K, Yan J. Antioxidant cascades confer neuroprotection in ethanol, morphine, and methamphetamine preconditioning. Neurochem Int 2019; 131:104540. [PMID: 31470038 DOI: 10.1016/j.neuint.2019.104540] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 08/06/2019] [Accepted: 08/23/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Shuang Lu
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China; Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Lvshuang Liao
- Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Bing Zhang
- School of Basic Medical Science, Xinjiang Medical University, Urumqi, 830001, China
| | - Weitao Yan
- Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Liangpei Chen
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - He Yan
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Limin Guo
- Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Shanshan Lu
- Histology and Embryology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Kun Xiong
- Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China.
| | - Jie Yan
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China; School of Basic Medical Science, Xinjiang Medical University, Urumqi, 830001, China.
| |
Collapse
|
16
|
Wang Y, Wu YP, Han JJ, Zhang MQ, Yang CX, Jiao P, Tian H, Zhu C, Qin SC, Sun XJ, Zhang HT, Zhao XM. Inhibitory effects of hydrogen on in vitro platelet activation and in vivo prevention of thrombosis formation. Life Sci 2019; 233:116700. [DOI: 10.1016/j.lfs.2019.116700] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/22/2019] [Accepted: 07/25/2019] [Indexed: 12/14/2022]
|
17
|
Wen D, Hui R, Wang J, Shen X, Xie B, Gong M, Yu F, Cong B, Ma C. Effects of Molecular Hydrogen on Methamphetamine-Induced Neurotoxicity and Spatial Memory Impairment. Front Pharmacol 2019; 10:823. [PMID: 31396089 PMCID: PMC6664236 DOI: 10.3389/fphar.2019.00823] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 06/25/2019] [Indexed: 12/17/2022] Open
Abstract
Methamphetamine (METH) is a highly addictive stimulant, and METH exposure can induce irreversible neuronal damage and cause neuropsychiatric and cognitive disorders. The ever-increasing levels of METH abuse worldwide have necessitated the identification of effective intervention strategies to protect the brain against METH-induced neurotoxicity. The protective effects of molecular hydrogen on oxidative stress and related neurodegenerative diseases have been recently elucidated. Herein, we investigated whether treatment with molecular hydrogen ameliorated the METH-induced neurotoxicity and spatial learning and memory impairments. Male C57BL/6 mice received four intraperitoneal METH injections (10 mg/kg, 3-h interval), and stereotypic behaviors and hyperthermia were observed. After METH treatment and behavioral observation, the mice were returned to their home cages, where they received water or hydrogen-rich water (HRW) ad libitum for 7 days. We found that the molecular hydrogen delivered by ad libitum HRW consumption significantly inhibited the METH-induced spatial learning impairment and memory loss evidenced in the Barnes maze and Morris water maze tests. Furthermore, molecular hydrogen significantly restrained the neuronal damage in the hippocampus after high-dose METH exposure. Ad libitum HRW consumption also had an inhibitory effect on the METH-induced increase in the expression of Bax/Bcl-2, cleaved caspase-3, glucose-related protein 78 (GRP 78), CCAAT/enhancer-binding protein homologous protein (CHOP), and p-NF-kB p65 expression and elevation of interleukin (IL)-6 and tumor necrosis factor (TNF)-α levels in the hippocampus. These are the first findings to indicate that hydrogen might ameliorate METH-induced neurotoxicity and has a potential application in reducing the risk of neurodegeneration frequently observed in METH abusers.
Collapse
Affiliation(s)
- Di Wen
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
| | - Rongji Hui
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
| | - Jian Wang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
| | - Xi Shen
- College of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Bing Xie
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
| | - Miao Gong
- Department of Histoembryology, Hebei Medical University, Shijiazhuang, China
| | - Feng Yu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
| | - Chunling Ma
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
| |
Collapse
|
18
|
Tan X, Shen F, Dong WL, Yang Y, Chen G. The role of hydrogen in Alzheimer's disease. Med Gas Res 2019; 8:176-180. [PMID: 30713672 PMCID: PMC6352568 DOI: 10.4103/2045-9912.248270] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 10/26/2018] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease is one of the most common neurodegenerative diseases in the elderly. It is often manifested as learning and memory impairment, cognitive function decline, normal social and emotional disorders. However, for this high-risk common disease, there is currently no effective treatment, which has plagued many clinicians. As a new type of medical therapeutic gas, hydrogen has attracted much attention recently. As a recognized reducing gas, hydrogen has shown great anti-oxidative stress and anti-inflammatory effect in many cerebral disease models. It can ameliorate neuronal damage, maintain the number of neurons, prolong the lifespan of neurons, and ultimately inhibit disease progression. Therefore, the role and mechanism of hydrogen in the pathological process of Alzheimer’s disease will be discussed in this paper.
Collapse
Affiliation(s)
- Xin Tan
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Fang Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Wan-Li Dong
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Yi Yang
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
19
|
Guo Q, Yin X, Qiao M, Jia Y, Chen D, Shao J, Lebaron TW, Gao Y, Shi H, Jia B. Hydrogen-Rich Water Ameliorates Autistic-Like Behavioral Abnormalities in Valproic Acid-Treated Adolescent Mice Offspring. Front Behav Neurosci 2018; 12:170. [PMID: 30127728 PMCID: PMC6087877 DOI: 10.3389/fnbeh.2018.00170] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/18/2018] [Indexed: 12/19/2022] Open
Abstract
Due to its anti-inflammatory and anti-oxidative effects, recent research has demonstrated that molecular hydrogen can serve as a new medical approach for depression, anxiety and traumatic brain injury. However, its potential effects on neurodevelopmental diseases, such as autism are still elusive. The present study aims to investigate the potential effects of hydrogen-rich water (HRW) administration on valproic acid (VPA)-induced autistic-like behavioral deficits, and the associated underlying mechanism in adolescent mice offspring. Pregnant ICR mice were randomly divided into five groups (n = 6). One group was injected with saline (NAV group) and provided hydrogen-free water. The other four groups were injected with VPA (600 mg/kg, intraperitoneally, i.p.) on pregnant day (PND) 12.5. One group was provided with hydrogen-free water (VEH group) and the other three groups were provided HRW at different segments, postnatal day 1 (PND 1) to PND 21 (PHV group), PND 13 to PND 21 (PVS group) or from PND 13 to postnatal day 42 (PVL group). Behavioral tests, including open field, novelty suppressed feeding (NSF), hot plate, social interaction (SI) and contextual fear memory tests were conducted between postnatal day 35–42. We found that HRW administration significantly reversed the autistic-like behaviors induced by maternal VPA exposure in the adolescent offspring of both male and female adolescent offspring. Furthermore, HRW administration significantly reversed the alternation of serum levels of interleukin 6 (IL-6) and tumor necrosis factor-α (TNF-α), but without any effects on the BDNF levels in maternal VPA-exposed mice offspring. These data suggest the need for additional research on HRW as a potential preventive strategy for autism and related disorders. Lay Summary: Maternal VPA injection induces autistic-like behavioral deficits in adolescent mice offspring. HRW administration ameliorates autistic-like behavioral deficits. HRW administration reverses the alternation of serum levels of IL-6 and TNF-α induced by VPA.
Collapse
Affiliation(s)
- Qingjun Guo
- Department of Surgery, Hebei Medical University, Shijiazhuang, China
| | - Xi Yin
- Department of Functional Region of Diagnosis, Hebei Medical University Fourth Hospital, Hebei Medical University, Shijiazhuang, China
| | - Meng Qiao
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Yujiao Jia
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Dandan Chen
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Juan Shao
- Department of Senile Disease, The Third Hospital of Hebei Medical University, Hebei Medical University, Shijiazhuang, China
| | | | - Yuan Gao
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang, China.,Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Haishui Shi
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang, China.,Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Forensic Medicine, Department of Forensic Medicine, Shijiazhuang, China.,Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medicial University, Shijiazhuang, China
| | - Bin Jia
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China.,Lingshui General Hospital, Lingshui, China
| |
Collapse
|
20
|
Abstract
Hydrogen is the most abundant chemical element in the universe, and has been used as an inert gas for a long time. More recent studies have shown that molecular hydrogen as a kind of antioxidant, anti-inflammatory, anti-apoptosis, gene expression and signal modulation molecule, can be used for the treatment of many diseases. This review mainly focuses on the research progresses of hydrogen in various medical fields and the possible action mechanisms.
Collapse
Affiliation(s)
- Hong-Mei Li
- Department of Cardiothoracic Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Li Shen
- Department of Cardiothoracic Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jun-Wen Ge
- Department of Cardiothoracic Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ru-Fang Zhang
- Department of Cardiothoracic Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|