1
|
Zare Z, Zarbakhsh S, Mohammadi M. Perinatal thyroid hormone deficiency leads to oxidative stress-induced neuronal damage and activation of astrocytes in rat hippocampus: Neuroprotective effect of exercise. Neuroscience 2025; 576:96-104. [PMID: 40300692 DOI: 10.1016/j.neuroscience.2025.04.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/11/2025] [Accepted: 04/24/2025] [Indexed: 05/01/2025]
Abstract
Thyroid hormones play a crucial role in proper brain development. This study examined the effects of developmental thyroid hormone deficiency on neuronal survival, glial fibrillary acidic protein (GFAP)-positive cells, and oxidative stress biomarkers in the hippocampus of congenital hypothyroid rats. The effectiveness of treadmill exercise in attenuating oxidative stress-induced neuronal damage and astrocyte activation was also evaluated. Pregnant Wistar rats in the hypothyroid group received propylthiouracil in their drinking water from gestational day 6 until weaning, while control dams received only tap water. Then, male offspring from both groups were further divided into two sub-groups: with or without four weeks of treadmill exercise. After sacrifice, the right hemispheres were processed for cresyl violet staining and immunohistochemistry, while the left hippocampi were used for biochemical assays. Results showed a reduced number of neurons and an increased number of GFAP-positive cells in the CA1 region of hypothyroid rats, with no significant changes in the CA3 region. Additionally, congenital hypothyroidism was associated with increased malondialdehyde levels, decreased glutathione levels, and reduced superoxide dismutase and catalase activity in the hippocampus. Treadmill exercise reduced astrocyte activation and protected neurons by inhibiting oxidative stress. Collectively, our results indicate that congenital thyroid hormone deficiency triggers astrocyte activation and compromises neuronal survival in the CA1 region by inducing oxidative stress. Exercise may serve as a beneficial supplementary treatment for attenuating oxidative stress-induced neuronal damage in congenital hypothyroidism.
Collapse
Affiliation(s)
- Zohreh Zare
- Department of Anatomical Sciences, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sam Zarbakhsh
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Moslem Mohammadi
- Department of Physiology, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
2
|
Pisani DF, Blondeau N. Deciphering the brain glucose metabolism: A gateway to innovative stroke therapies. J Cereb Blood Flow Metab 2025:271678X251346277. [PMID: 40439074 PMCID: PMC12122490 DOI: 10.1177/0271678x251346277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 05/14/2025] [Accepted: 05/14/2025] [Indexed: 06/02/2025]
Abstract
Stroke is the leading cause of physical disability and death among adults in most Western countries. Consecutive to a vascular occlusion, cells face a brutal reduction in supply of oxygen and glucose and thus an energy failure, which in turn triggers cell death mechanisms. Among brain cells, neurons are the most susceptible to ischemia because of their high metabolic demand and low reservoir of energy substrates. In neurons, glycolysis uses glucose coming from blood or from glycogen stored in astrocytes, underlying the deep astrocyte-neuron metabolic cooperation. During ischemia, both the aerobic and anaerobic pathways and thus energy production are compromised, which disrupts proper cell functioning, notably Na+/K+ ATPase and mitochondria. This results in altered Ca2+ homeostasis and overproduction of ROS, the latter being further exacerbated during the reperfusion phase. Consequently, glucose metabolism in the different brain cell populations plays a central role in injury and recovery after stroke, and has recently emerged as a promising target for therapeutic intervention. In this context, the overall objective of this article is to review the interconnections between stroke and brain glucose metabolism and to explore how its targeting may offer new therapeutic opportunities in addressing the global stroke epidemic.
Collapse
Affiliation(s)
- Didier F Pisani
- Université Côte d’Azur, CNRS, LP2M, Nice, France
- Laboratories of Excellence Ion Channel Science and Therapeutics, Nice, France
| | - Nicolas Blondeau
- Laboratories of Excellence Ion Channel Science and Therapeutics, Nice, France
- Université Côte d’Azur, CNRS, Inserm, IPMC, Valbonne, France
| |
Collapse
|
3
|
Wang C, Feng H, Zhao J, Liu C, Wang X, Zhou H, Mai K, He G. Oxidative stress and metabolic perturbations unravel the molecular basis of high dietary poultry by-product meal-induced growth impairment and inflammation response in Litopenaeusvannamei. FISH & SHELLFISH IMMUNOLOGY 2025; 164:110433. [PMID: 40404030 DOI: 10.1016/j.fsi.2025.110433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 04/26/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025]
Abstract
The pacific white shrimp, Litopenaeus vannamei (L. vannamei) is one of the most widely farmed shrimp species in global aquaculture. The scarcity of fishmeal as a component of aquafeeds poses a considerable challenge to the sustainable development of the aquaculture industry. This study aimed to assess the feasibility of poultry by-product meal (PBM) as an alternative to fishmeal of L. vannamei diets by assessing its impact on growth performance, immune response, postprandial metabolism and signaling pathways. Six diets were formulated to be iso-nitrogenous (42 %) and iso-lipidic (7 %), with 0, 8 %, 16 %, 25 %, 33 %, and 50 % fishmeal replaced by poultry by-product meal (marked as CON, PBM8, PBM16, PBM25, PBM33, PBM50). After an 8 weeks of growth trial, results showed that dietary PBM replacement up to 16 % didn't negatively affect growth performance of L. vannamei. However, PBM replacement at 16 % or higher led to significant reductions in lipase and trypsin activity, deteriorated gut morphology, and disrupted the balance of immune-related gene expression and phosphorylation of Nf-κB in the intestine or hepatopancreas. Mechanically, oxidative stress progressively increased in the hepatopancreas with higher dietary PBM levels, inducing ferroptosis and cuproptosis while leaving apoptosis and autophagy unaffected. Postprandial mTORC1 signaling in muscle was significantly suppressed by higher PBM levels, impairing protein synthesis. Furthermore, high PBM replacement disrupted postprandial energy metabolism, affecting glycolysis, gluconeogenesis, and TCA cycle pathways. Taken together, these results shed light on the molecular, immune and metabolic impacts of dietary PBM replacement in L. vannamei, enhancing comprehension of the relationship between dietary protein sources and shrimp physiological health.
Collapse
Affiliation(s)
- Chenxi Wang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China
| | - Huali Feng
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China
| | - Jican Zhao
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China
| | - Chengdong Liu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China.
| | - Xuan Wang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China
| | - Huihui Zhou
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China
| | - Kangsen Mai
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China
| | - Gen He
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China; Key Laboratory of Aquaculture Nutrition and Feeds, Ministry of Agriculture, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
4
|
Zhao Q, Hao D, Wang S, Chen S, Zhou C, Fan C, Su Q, Huang W, Liu J, Kong Q, Wu Y, He Z. Exposure to high altitude leads to disturbances in host metabolic homeostasis: study of the effects of hypoxia-reoxygenation and the associations between the microbiome and metabolome. mSystems 2025; 10:e0134724. [PMID: 40237534 PMCID: PMC12090774 DOI: 10.1128/msystems.01347-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/16/2025] [Indexed: 04/18/2025] Open
Abstract
This study investigated alterations in hematological parameters, gut microbiota composition, and fecal and plasma metabolic profiles among high-altitude residents during reoxygenation periods of 1 week, 1 month, and 4 months to elucidate the effects of reoxygenation on human physiology and metabolism. Exposure to high altitudes alters intestinal flora, plasma and fecal metabolites, disrupting their metabolic balance. Distinct differences in amino acid, lipid, energy, immune, cofactor, and vitamin metabolism pathways were detected between high- and low-altitude populations, with a partial recovery of disparities during reoxygenation. Although the gut microbiota exhibited limited adaptive homeostasis to altitude variations, the abundance of microbial taxa and the expression levels of fecal metabolites during the initial reoxygenation phase, particularly during the first week, were sensitive to the reoxygenated environment. Through 16S rRNA gene sequencing and bioinformatics analysis, operational taxonomic units (OTUs) were annotated at the genus level, revealing that the genera Barnesiella, Parabacteroides, and Megasphaera, along with plasma L-arginine, S1P, and alpha-D-glucose, emerged as potential biomarkers for the first week of reoxygenation among high-altitude populations. Notably, a marked change in oxidative stress levels and an increase in antioxidant capacity were observed in high-altitude residents during early reoxygenation. Tyrosine metabolism, which is jointly regulated by the plasma and fecal metabolites and gut microbiota, plays an important role under high-altitude conditions during initial reoxygenation. Additionally, the plasma metabolites pyridoxine and hypoxanthine and the Rothia genus correlated significantly with high-altitude deacclimatization syndrome scores during the first week of reoxygenation.IMPORTANCEOur research focuses on the prompt activation of tyrosine metabolism in plasma following reoxygenation, along with the regulatory mechanisms employed by the intestinal microbiota and the metabolism of feces to modulate this metabolic process. Notably, in the initial stages of reoxygenation, specific microbial genera such as Barnesiella, Parabacteroides, and Megasphaera, alongside plasma biomarkers including L-arginine, S1P, and alpha-D-glucose, emerge as pivotal players. Additionally, our findings reveal a distinct hematological profile characterized by a decrease in the MCHC and increases in the MCV and RDW-SD during the first week of reoxygenation, and this temporal window marked a crucial juncture in the plasma metabolome. Whereas the first month of reoxygenation signified a pivotal phase in the gut microbiome's adaptation to altered environmental conditions, as evidenced by alterations in alpha diversity.
Collapse
Affiliation(s)
- Qin Zhao
- Department of Biobank, Hospital of Chengdu Office of People’s Government of Xizang Autonomous Region (Hospital.C.X.), Chengdu, Sichuan, China
| | - Doudou Hao
- Department of Biobank, Hospital of Chengdu Office of People’s Government of Xizang Autonomous Region (Hospital.C.X.), Chengdu, Sichuan, China
| | - Siyu Wang
- Department of Biobank, Hospital of Chengdu Office of People’s Government of Xizang Autonomous Region (Hospital.C.X.), Chengdu, Sichuan, China
| | - Siyuan Chen
- Department of Biobank, Hospital of Chengdu Office of People’s Government of Xizang Autonomous Region (Hospital.C.X.), Chengdu, Sichuan, China
| | - Chaohua Zhou
- Department of Biobank, Hospital of Chengdu Office of People’s Government of Xizang Autonomous Region (Hospital.C.X.), Chengdu, Sichuan, China
| | - Chen Fan
- Department of Science Education, Hospital of Chengdu Office of People’s Government of Xizang Autonomous Region (Hospital.C.X.), Chengdu, Sichuan, China
| | - Qian Su
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Wenting Huang
- Stomatology, Hospital of Chengdu Office of People’s Government of Xizang Autonomous Region (Hospital.C.X.), Chengdu, Sichuan, China
| | - Jiaxin Liu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, Sichuan, China
| | - Qingquan Kong
- Department of Orthopedics, Hospital of Chengdu Office of People’s Government of Xizang Autonomous Region (Hospital.C.X.), Chengdu, Sichuan, China
| | - Yunhong Wu
- Department of Endocrinology and Metabolism, Hospital of Chengdu Office of People’s Government of Xizang Autonomous Region (Hospital.C.X.), Chengdu, Sichuan, China
| | - Zeng He
- Department of Biobank, Hospital of Chengdu Office of People’s Government of Xizang Autonomous Region (Hospital.C.X.), Chengdu, Sichuan, China
| |
Collapse
|
5
|
Alves JL, Quinta-Ferreira RM, Quinta-Ferreira ME, Matias CM. Exploring different mechanisms of reactive oxygen species formation in hypoxic conditions at the hippocampal CA3 area. Mol Cell Endocrinol 2025; 601:112517. [PMID: 40054836 DOI: 10.1016/j.mce.2025.112517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/17/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Hypoxia can lead to severe consequences for brain function, particularly in regions with high metabolic demands such as the hippocampus. Excessive production of reactive oxygen species (ROS) during hypoxia can initiate a cascade of oxidative stress, evoking cellular damage and neuronal dysfunction. Most of the studies characterizing the formation of ROS are performed in the context of ischemia induced by oxygen-glucose deprivation, thus, the role of hypoxia in less severe conditions requires further clarification. The aim of this work was to identify the major mechanisms of ROS generation and assess flavoprotein autofluorescence changes. For ROS detection, the slices were incubated with the indicator H2DCFDA, while intrinsic FAD-linked autofluorescence was recorded from indicator free slices. All signals were measured under hypoxia, at the hippocampal mossy fiber synapses of CA3 area, which were chemically stimulated using 20 mM KCl. The results suggest that ROS is formed in the mitochondria, during moderate hypoxia. The blockage of mitochondrial complexes I, III and IV with rotenone, myxothiazol and sodium azide, respectively, and of the mitochondrial calcium uniporter with Ru265, led to the abolishment of ROS changes and to an increase of FAD-linked autofluorescence (with the exception of the complexes III and IV). The blockage of the enzyme oxidases NADPH and xanthine oxidase also impaired ROS formation and rose FAD-linked autofluorescence. Thus, the blockage of any of the steps of the process of ROS formation, namely the activation of critical MRC complexes, calcium entry into the mitochondria, or enzyme oxidases activity, ceases the production of ROS.
Collapse
Affiliation(s)
- João L Alves
- Department of Life Sciences, University of Coimbra, Portugal; CNC-UC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal
| | - Rosa M Quinta-Ferreira
- CERES - Chemical Engineering and Renewable Resources for Sustainability, Department of Chemical Engineering, University of Coimbra, Portugal
| | - M Emília Quinta-Ferreira
- CNC-UC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal; Department of Physics, University of Coimbra, Portugal
| | - Carlos M Matias
- CNC-UC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal; Department of Physics, UTAD, Vila Real, Portugal.
| |
Collapse
|
6
|
Yang C, Tang S, Liu Q, Fan M, Zhang W, Liu Y, Chen X, Xu G, Chen X, Xu Z. Wireless charging LED mediated type I photodynamic therapy of breast cancer using NIR AIE photosensitizer. iScience 2025; 28:112196. [PMID: 40230527 PMCID: PMC11995052 DOI: 10.1016/j.isci.2025.112196] [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: 01/12/2025] [Revised: 02/09/2025] [Accepted: 03/06/2025] [Indexed: 04/16/2025] Open
Abstract
Due to limited light penetration and dependence on oxygen, photodynamic therapy (PDT) is typically restricted to treating shallow tissues. Developing strategies to overcome these limitations and effectively using PDT for tumor treatment is a significant yet unresolved challenge. In this study, we present a smart approach combining a wireless-charged LED (wLED) with a type I aggregation-induced emission photosensitizer, MeOTTMN, to address both light penetration and tumor hypoxia issues simultaneously. MeOTTMN, characterized by twisted molecular architecture and strong intramolecular electron donor-acceptor interaction, produces high levels of hydroxyl and superoxide radicals and emits near-infrared light in its aggregated state, thus facilitating fluorescence imaging-guided PDT once formulated into nanoparticles. The inhibition of breast cancer xenografts provides compelling evidence of the treatment efficacy of type I PDT irradiated through an implantable wLED. This strategy provides a conceptual and practical paradigm to overcome key clinical limitations of PDT, expanding possibilities for clinical translation.
Collapse
Affiliation(s)
- Chengbin Yang
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Shiqi Tang
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Qiqi Liu
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Miaozhuang Fan
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Wenguang Zhang
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Yingyu Liu
- Maternal-Fetal Medicine Institute, Department of Obstetics and Gynaecology, Shenzhen Baoan Women’s and Children’s Hospital, Shenzhen 518133, China
| | - Xin Chen
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Gaixia Xu
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Xiaoyan Chen
- Maternal-Fetal Medicine Institute, Department of Obstetics and Gynaecology, Shenzhen Baoan Women’s and Children’s Hospital, Shenzhen 518133, China
| | - Zhourui Xu
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| |
Collapse
|
7
|
Ibrahim E, Sohail SK, Ihunwo A, Eid RA, Al-Shahrani Y, Rezigalla AA. Effect of high-altitude hypoxia on function and cytoarchitecture of rats' liver. Sci Rep 2025; 15:12771. [PMID: 40229399 PMCID: PMC11997024 DOI: 10.1038/s41598-025-97863-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 04/08/2025] [Indexed: 04/16/2025] Open
Abstract
The liver is central to metabolic, detoxification, and homeostatic functions. Exposure to hypobaric hypoxia at high altitudes causes detrimental effects on the liver, leading to injury. This study evaluated the effect of hypoxia-induced at high altitudes on liver function, oxidative stress, and histopathological changes in rats. This study used 24 male Wistar rats (aged 8-10 weeks). The hypoxia (hypobaric hypoxia) was inducted at a high altitude of 2,100 m above sea level. Normoxia is defined as 40 m above the sea level. The rats were randomly divided into two groups: a control group maintained at low altitudes and an experimental group exposed to high altitudes for eight weeks. Blood samples were collected from all rats through a cardiac puncture, and liver samples were taken through an abdominal approach. All samples were processed through standard methods and evaluated for liver function tests and histopathological assessment. Serum aspartate aminotransferase and alanine transaminase levels significantly increased by 25% and 30%, respectively, in the high-altitude group compared to controls (p < 0.01), indicating mild hepatocellular damage. Oxidative stress assessment indicated a significant elevation in malondialdehyde by 42% in the liver homogenates of high-altitude rats compared to controls (p < 0.001). Moreover, Superoxide dismutase activity and glutathione content decreased by 18% and 22% in the high-altitude group (p < 0.01), confirming the increased oxidative stress. Histologically, minimal inflammatory infiltration was observed in the rat livers at high altitudes, with no signs of necrosis or severe structural changes. Subclinical liver dysfunction, as evidenced by altered serum enzyme levels and increased oxidative stress with mild histological changes, is induced by high-altitude hypoxia in rats. This study's results support that a hypobaric hypoxic environment physiologically stresses the liver. Further research into the long-term implications of hypobaric hypoxia and the adaptive responses of the liver is warranted.
Collapse
Grants
- UB-14-1442 Deputyship for Research & Innovation, Ministry of Education, in Saudi Arabia, which has supported this research work with the project number (UB-14-1442).
- UB-14-1442 Deputyship for Research & Innovation, Ministry of Education, in Saudi Arabia, which has supported this research work with the project number (UB-14-1442).
- UB-14-1442 Deputyship for Research & Innovation, Ministry of Education, in Saudi Arabia, which has supported this research work with the project number (UB-14-1442).
- UB-14-1442 Deputyship for Research & Innovation, Ministry of Education, in Saudi Arabia, which has supported this research work with the project number (UB-14-1442).
- UB-14-1442 Deputyship for Research & Innovation, Ministry of Education, in Saudi Arabia, which has supported this research work with the project number (UB-14-1442).
- UB-14-1442 Deputyship for Research & Innovation, Ministry of Education, in Saudi Arabia, which has supported this research work with the project number (UB-14-1442).
- Deputyship for Research & Innovation, Ministry of Education, in Saudi Arabia, which has supported this research work with the project number (UB-14-1442).
Collapse
Affiliation(s)
- Elwathiq Ibrahim
- Department of Anatomy, College of Medicine, University of Bisha, Bisha, 61922, Saudi Arabia
| | - Shahzada Khalid Sohail
- Department of Pathology, College of Medicine, University of Bisha, Bisha, 61922, Saudi Arabia
| | - Amadi Ihunwo
- School of Anatomical Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Refaat A Eid
- Department of Pathology, College of Medicine, King Khalid University, Abha, 62529, 12573, Saudi Arabia
| | - Yazeed Al-Shahrani
- Department of Emergency Medicine, King Abdalla Hospital, Health Affairs Administration, Bisha, Saudi Arabia
| | - Assad Ali Rezigalla
- Department of Anatomy, College of Medicine, University of Bisha, Bisha, 61922, Saudi Arabia.
| |
Collapse
|
8
|
Yadav S, Singh S, Singh M. Protective effects of Tanshinone IIA preconditioning against hypobaric hypoxia-induced lung injury in a rat model. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04118-7. [PMID: 40227308 DOI: 10.1007/s00210-025-04118-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025]
Abstract
Tanshinone IIA (Tan-IIA), derived from Salvia miltiorrhiza, has been used in traditional Chinese medicine to treat cardiovascular diseases and pulmonary hypertension. This study investigates the potential of Tan-IIA preconditioning as a protective strategy against hypoxia-induced lung injury. Male Sprague-Dawley rats (200 ± 25 g) were divided into four groups: normoxia, normoxia with Tan-IIA, hypobaric hypoxia, and hypobaric hypoxia with Tan-IIA. Tan-IIA was administered intraperitoneally at doses of 10, 20, and 40 mg/kg body weight one hour before exposure to hypobaric hypoxia (simulated altitude of 25,000 feet for 48 h). Pulmonary edema was assessed by measuring transvacuolar leakage of sodium fluorescein dye and lung water content. Exposure to hypoxia triggered redox imbalances, inflammation, and changes in levels of nitric oxide (NOx), endothelin- 1, and Na/K ATPase, which contributed to pulmonary edema. Tan-IIA preconditioning, particularly at 20 mg/kg, was effective in reversing these disturbances. Tan-IIA modulated the expression of key signaling molecules, including c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK), and p38, as well as downstream activator protein- 1 (AP- 1) subunits (Jun and Fos), thus reducing inflammation. Its protective effects were partly due to increased NO levels and decreased endothelin, which lowered pulmonary vasoconstriction and permeability, respectively. Additionally, enhanced Na/K ATPase expression via hypoxia-inducible factor- 1 (HIF- 1) and AP- 1 pathways promoted alveolar fluid clearance, while interactions between nuclear factor erythroid 2-related factor 2 (Nrf2) and c-Jun highlighted the anti-oxidative properties of Tanshinone IIA. These findings demonstrate that Tanshinone IIA preconditioning protects against hypoxia-induced lung injury by mitigating pulmonary leakage. This highlights its potential therapeutic application in hypoxic lung conditions.
Collapse
Affiliation(s)
- Seema Yadav
- Experimental Biology Division, Defence Institute of Physiology and Allied Science, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110 054, India
| | - Somnath Singh
- Experimental Biology Division, Defence Institute of Physiology and Allied Science, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110 054, India
| | - Mrinalini Singh
- Experimental Biology Division, Defence Institute of Physiology and Allied Science, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110 054, India.
| |
Collapse
|
9
|
Chen J, Zhang Y, Jiang C, Chen S, Zhao Y, Gao X, Guo X, Hu G, Liu P, Jin H, Zhang Y, Omar SM, Li L, Wan G, Liu P. Dichlorvos poisoning caused chicken cerebellar autophagy and changes of Caecal microflora. Poult Sci 2025; 104:104939. [PMID: 40068572 PMCID: PMC11932683 DOI: 10.1016/j.psj.2025.104939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
In order to explore the effects of acute dichlorvos exposure on cerebellar autophagy and cecal microbes in broilers and to analyze the relationship between autophagy-related genes and cecal microbes. Broilers were randomly divided into three groups (with 16 broilers in each group)and respectively given distilled water and dichlorvos (2.48 mg / kg, 11.3 mg / kg). The cecal contents and cerebellum samples were collected after poisoning symptoms of broilers, and the antioxidant indexes such as SOD and CAT in cerebellum were detected. Hematoxylin-eosin (HE) staining of cerebellum and cecum, and immunofluorescence sections of cerebellum LC3 were made. RT-PCR and western blot were used to detect the expression of oxidative stress and autophagy-related genes in cerebellar tissue. The cecal contents were analyzed by 16S rRNA high-throughput sequencing, and then the correlation between the expression of autophagy-related genes and the abundance of intestinal microbes was analyzed. It was concluded that dichlorvos exposure destroyed the normal morphological structure of the cerebellum and cecum in broilers, which induced oxidative stress and autophagy in the cerebellum of broilers, reduced the diversity of cecal microorganisms, and destroyed the steady state of the cecal microbial structure. In addition, The changes of mRNA expression of autophagy-related genes is related to some specific bacteria. In summary, this study found that dichlorone exposure can cause cerebellar oxidative stress and autophagy, and the mechanism of cerebellar injury in broilers is linked to cecal microbiota changes, potentially offering a new direction for researching dichlorone's pathogenic mechanism.
Collapse
Affiliation(s)
- Juan Chen
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, PR China
| | - Yun Zhang
- Huaihua City Maternal and Child Health Care Hospital, Huaihua, Hunan 418000, PR China
| | - Chenxi Jiang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, PR China
| | - Shupeng Chen
- Jiangxi Agricultural Engineering Vocational college, Zhangshu, Jiangxi 331200, PR China
| | - Yulan Zhao
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, PR China
| | - Xiaona Gao
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, PR China
| | - Xiaoquan Guo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, PR China
| | - Guoliang Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, PR China
| | - Pei Liu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, PR China
| | - Huibo Jin
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, PR China
| | - Ying Zhang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, PR China
| | - Salma Mbarouk Omar
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, PR China
| | - Lin Li
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, PR China
| | - Gen Wan
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, PR China
| | - Ping Liu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, PR China.
| |
Collapse
|
10
|
Luo L, Zhang S, Gong J, Zhang J, Xie P, Yin J, Zhang M, Zhang C, Chen H, Liu Y, Ni B, Li C, Tian Z. 3-D Sustained-Release Culture Carrier Alleviates Rat Intervertebral Disc Degeneration by Targeting STING in Transplanted Skeletal Stem Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410151. [PMID: 39985222 PMCID: PMC12005824 DOI: 10.1002/advs.202410151] [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: 08/23/2024] [Revised: 01/14/2025] [Indexed: 02/24/2025]
Abstract
The hypoxic and high-pressure microenvironment of the intervertebral discs poses a major challenge to the survival and therapeutic efficiency of exogenous stem cells. Therefore, improving the utilization efficiency and therapeutic effect of exogenous stem cells to delay intervertebral disc degeneration (IVDD) is of great importance. Here, hypoxic induction studies are conducted in vivo and in vitro using rat costal cartilage-derived skeletal stem cells (SSCs) and find that hypoxia activates the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)/stimulator of interferon genes (STING) signaling pathway and increased reactive oxygen species (ROS) accumulation, triggering ferroptosis in SSCs through hypoxia-inducible factor-1 alpha-dependent mitophagy. Progressive hypoxia preconditioning reduce STING expression and ROS accumulation, inducing SSCs differentiation into nucleus pulposus-like cells via the Wnt signaling pathway. Considering this, a 3-D sustained-release culture carrier is generated by mixing SSCs with methacrylated hyaluronic acid and polydopamine nanoparticles coated with the STING inhibitor C-176 and evaluated its inhibitory effect on IVDD. This carrier is demonstrated to inhibit the cGAS/STING pathway and prevent ROS accumulation by continuously releasing C-176-coated polydopamine nanoparticles, thereby reducing ferroptosis, promoting differentiation, and ultimately attenuating IVDD, suggesting its potential as a novel treatment strategy.
Collapse
Affiliation(s)
- Liwen Luo
- Department of OrthopaedicsXinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingP. R. China
- State Key Laboratory of Trauma and Chemical PoisoningArmy Medical University (Third Military Medical University)ChongqingP. R. China
| | - Shiyu Zhang
- Department of OrthopaedicsXinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingP. R. China
| | - Junfeng Gong
- Department of General SurgeryThe Armed Police Corps Hospital of AnhuiHefeiP. R. China
| | - Ji Zhang
- Institute of ImmunologyPLAArmy Medical University (Third Military Medical University)ChongqingP. R. China
| | - Peng Xie
- Department of Military BiosafetyCollege of Basic MedicineArmy Medical UniversityChongqingP. R. China
| | - Jun Yin
- Department of PathophysiologyCollege of High Altitude Military MedicineArmy Military Medical UniversityChongqingP. R. China
| | - MengJie Zhang
- Department of PathophysiologyCollege of High Altitude Military MedicineArmy Military Medical UniversityChongqingP. R. China
| | - Cong Zhang
- Department of Laboratory Animal ScienceCollege of Basic MedicineArmy Medical UniversityChongqingP. R. China
| | - Hong Chen
- Department of Orthopedics903 Hospital of Joint Logistic Support Force of The People's Liberation ArmyHangzhouP. R. China
| | - Yao Liu
- Department of PharmacyDaping HospitalArmy Medical University (Third Military Medical University)ChongqingP. R. China
| | - Bing Ni
- Department of PathophysiologyCollege of High Altitude Military MedicineArmy Military Medical UniversityChongqingP. R. China
| | - Changqing Li
- Department of OrthopaedicsXinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingP. R. China
- State Key Laboratory of Trauma and Chemical PoisoningArmy Medical University (Third Military Medical University)ChongqingP. R. China
| | - Zhiqiang Tian
- Institute of ImmunologyPLAArmy Medical University (Third Military Medical University)ChongqingP. R. China
| |
Collapse
|
11
|
Lu G, Rili G, Shuang M. Impact of hypoxia on the hippocampus: A review. Medicine (Baltimore) 2025; 104:e41479. [PMID: 40128035 PMCID: PMC11936607 DOI: 10.1097/md.0000000000041479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 01/20/2025] [Indexed: 03/26/2025] Open
Abstract
Oxygen is the most abundant chemical substance and is a basic material for human activities. A decline in oxygen concentration affects many physiological processes in the body, leading to pathological changes and even the occurrence of diseases. Therefore, an increasing number of studies have focused on the pathological state of hypoxia. The hippocampus is the most sensitive tissue to oxygen in the brain. The reduction in oxygen concentration affects the morphology and functioning of the hippocampus, including a decline in learning and memory, immunity, and energy metabolism, causing great problems to people's physical and mental health. To keep people healthy in hypoxic environments, adapt to hypoxic environments, and avoid diseases, it is necessary to review the morphology and function of the hippocampus, as well as the effect of oxygen on the hippocampus.
Collapse
Affiliation(s)
- Guan Lu
- Research Center for High Altitude Medicine, School of Medicine, Qinghai University, Xining, China
- Key Laboratory for Application for High Altitude Medicine, Qinghai University, Xining, China
| | - Ge Rili
- Research Center for High Altitude Medicine, School of Medicine, Qinghai University, Xining, China
- Key Laboratory for Application for High Altitude Medicine, Qinghai University, Xining, China
| | - Ma Shuang
- Research Center for High Altitude Medicine, School of Medicine, Qinghai University, Xining, China
- Key Laboratory for Application for High Altitude Medicine, Qinghai University, Xining, China
| |
Collapse
|
12
|
Poetzsch G, Jelacic L, Dammer L, Hellmann SL, Balling M, Andrade-Navarro M, Avivi A, Shams I, Bicker A, Hankeln T. Adaptation of the Spalax galili transcriptome to hypoxia may underlie the complex phenotype featuring longevity and cancer resistance. NPJ AGING 2025; 11:16. [PMID: 40044716 PMCID: PMC11882797 DOI: 10.1038/s41514-025-00206-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025]
Abstract
In the subterranean rodent (Nanno)spalax galili, evolutionary adaptation to hypoxia is correlated with longevity and tumor resistance. Adapted gene-regulatory networks of Spalax might pinpoint strategies to maintain health in humans. Comparing liver, kidney and spleen transcriptome data from Spalax and rat at hypoxia and normoxia, we identified differentially expressed gene pathways common to multiple organs in both species. Body-wide interspecies differences affected processes like cell death, antioxidant defense, DNA repair, energy metabolism, immune response and angiogenesis, which may play a crucial role in Spalax's adaptation to environmental hypoxia. In all organs, transcription of genes for genome stability maintenance and DNA repair was elevated in Spalax versus rat, accompanied by lower expression of aerobic energy metabolism and proinflammatory genes. These transcriptomic changes might account for the extraordinary lifespan of Spalax and its cancer resistance. The identified gene networks present candidates for further investigating the molecular basis underlying the complex Spalax phenotype.
Collapse
Affiliation(s)
- Gesa Poetzsch
- Molecular Genetics & Genome Analysis, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg-University, Mainz, Germany
| | - Luca Jelacic
- Molecular Genetics & Genome Analysis, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg-University, Mainz, Germany
| | - Leon Dammer
- Molecular Genetics & Genome Analysis, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg-University, Mainz, Germany
| | - Sören Lukas Hellmann
- Molecular Genetics & Genome Analysis, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg-University, Mainz, Germany
- Nucleic Acids Core Facility, Faculty of Biology, Johannes Gutenberg-University, Mainz, Germany
| | - Michelle Balling
- Molecular Genetics & Genome Analysis, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg-University, Mainz, Germany
| | - Miguel Andrade-Navarro
- Computational Biology and Data Mining Group, Institute of Organismic and Molecular Evolution, Johannes Gutenberg University, Mainz, Germany
| | - Aaron Avivi
- Institute of Evolution, University of Haifa, Mount Carmel, Haifa, Israel
| | - Imad Shams
- Institute of Evolution, University of Haifa, Mount Carmel, Haifa, Israel
- Department of Evolutionary and Environmental Biology, Faculty of Natural Sciences, University of Haifa, Mount Carmel, Haifa, Israel
| | - Anne Bicker
- Molecular Genetics & Genome Analysis, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg-University, Mainz, Germany
- Department of Medicine I, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Thomas Hankeln
- Molecular Genetics & Genome Analysis, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg-University, Mainz, Germany.
| |
Collapse
|
13
|
Zhang Q, Xu Y, Luo H, Su H, Zhong J, Pan L, Liu Y, Yang C, Yin Y, Tan B. Treadmill Training-Induced Remyelination Rescues Cognitive Impairment After Acute Hypoxia. Neurochem Res 2025; 50:109. [PMID: 40025348 DOI: 10.1007/s11064-025-04359-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/24/2024] [Accepted: 02/14/2025] [Indexed: 03/04/2025]
Abstract
Acute and chronic exposure to high altitude causes multiple negative neurological consequences. Further research has shown the efficacy of targeted drugs after acute hypoxia. However, the effects and mechanisms of physical therapy like exercise, on after exposed-induced myelin repair and functional improvements have remained unclear. Here, we explored the efficacy of treadmill training at different intensities on recovery in a rat model of acute hypobaric hypoxia (HH) injury. A 4-week treadmill training scheme was used at 30%, 50%, and 70% of maximum speed. The evolution of oligodendrocyte morphometry was observed by immunofluorescence, and the expressions of myelin-related proteins were detected by western blotting. Transmission electron microscopy (TEM) is used to study fine myelin structure. In addition, the open field test (OFT), elevated plus maze (EPM) and Morris water maze (MWM) were used for the observation of cognitive function recovery. Our study revealed varying degrees of demyelination changes in the cortex and hippocampus following acute hypoxia exposure. Additionally, high-intensity treadmill training enhances oligodendrocyte (OL) maturation, improves myelin-related proteins, and increases myelin sheath thickness, thus facilitating myelin repair, rescuing cognitive function and mood disorders, and preserving normal nerve conduction. Finally, the upregulation of peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC1α) and key enzymes of cholesterol synthesis (HMGCR/FDPS) induced by high-intensity treadmill training was detected. Our results demonstrate that high-intensity treadmill training as a physical therapy via PGC1α and cholesterol synthesis enhances myelin repair and functional restoration, which should provide new insight for the rehabilitation of remyelination by exercise.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Chongqing, 400000, China
| | - Yangjie Xu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Chongqing, 400000, China
| | - Haodong Luo
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Chongqing, 400000, China
| | - Hong Su
- Guangzhou Women and Children'S Medical Center, Guangzhou Medical University, Guangzhou, 510620, China
| | - Juan Zhong
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Chongqing, 400000, China
| | - Lu Pan
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Chongqing, 400000, China
| | - Yuan Liu
- State Key Laboratory of Trauma, Burns and Combined Injuries, Department of Special Environment War Wound Prevention and Treatment, Institute of Surgery Research, Army Medical Center of PLA, Chongqing, 400000, China
| | - Ce Yang
- State Key Laboratory of Trauma, Burns and Combined Injuries, Department of Special Environment War Wound Prevention and Treatment, Institute of Surgery Research, Army Medical Center of PLA, Chongqing, 400000, China
| | - Ying Yin
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Chongqing, 400000, China.
| | - Botao Tan
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Chongqing, 400000, China.
| |
Collapse
|
14
|
Dickmeiß J, Henning Y, Stahlke S, Weber T, Theiss C, Matschke V. Differential Protective Effects of Edaravone in Cerebellar and Hippocampal Ischemic Injury Models. CEREBELLUM (LONDON, ENGLAND) 2025; 24:49. [PMID: 39964549 PMCID: PMC11835913 DOI: 10.1007/s12311-025-01804-3] [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] [Accepted: 02/11/2025] [Indexed: 02/21/2025]
Abstract
Ischemic stroke is a leading cause of mortality and disability, with cerebellar strokes posing severe complications such as herniation and brainstem compression. Edaravone, a radical scavenger known for reducing oxidative stress, has shown neuroprotective effects in cerebral strokes, but its impact on cerebellar strokes remains unclear. This study investigates Edaravone's protective properties in organotypic slice cultures of rat cerebellum and hippocampus, employing an oxygen-glucose deprivation (OGD) model to simulate ischemic stroke. The hippocampus served as comparative structure due to its high hypoxia sensitivity. Our results confirmed effective hypoxic induction with increases in HIF-1α and HIF-2α expression. Edaravone significantly reduced lactate dehydrogenase (LDH) levels, indicating diminished cellular damage, with cerebellar tissues showing greater vulnerability. Additionally, Edaravone reduced reactive oxygen species (ROS) in both tissues, though its efficacy may be limited by higher oxidative stress in cerebellar cultures. Seahorse XF analysis revealed that Edaravone preserved mitochondrial respiration and tissue integrity in cerebellar and hippocampal slice cultures. However, Edaravone was more effective in preserving mitochondrial respiration in hippocampal slices, suggesting that OGD-induced damage is more severe in cerebellar tissue. In conclusion, Edaravone demonstrates significant cell protective effects in both cerebellar and hippocampal tissues under OGD conditions, preserving tissue integrity and enhancing mitochondrial function in a tissue-dependent manner. These findings suggest Edaravone as a promising therapeutic candidate for cerebellar stroke. Further in vivo studies are required to assess its full clinical potential.
Collapse
Affiliation(s)
- Jens Dickmeiß
- Department of Cytology, Institute of Anatomy, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Yoshiyuki Henning
- Institute of Physiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sarah Stahlke
- Department of Cytology, Institute of Anatomy, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Thomas Weber
- Department of Anesthesiology and Intensive Care Medicine, Ruhr University Bochum, St. Josef Hospital, D-44791, Bochum, Germany
| | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
- International Graduate School of Neuroscience (IGSN), Ruhr-University Bochum, 44801, Bochum, Germany
| | - Veronika Matschke
- Department of Cytology, Institute of Anatomy, Medical Faculty, Ruhr-University Bochum, Bochum, Germany.
- International Graduate School of Neuroscience (IGSN), Ruhr-University Bochum, 44801, Bochum, Germany.
| |
Collapse
|
15
|
Bambor C, Daunheimer S, Raffort C, Koedel J, Salameh A, Raßler B. Effects of a Three-Day vs. Six-Day Exposure to Normobaric Hypoxia on the Cardiopulmonary Function of Rats. Curr Issues Mol Biol 2025; 47:125. [PMID: 39996846 PMCID: PMC11854188 DOI: 10.3390/cimb47020125] [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: 01/08/2025] [Revised: 02/04/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025] Open
Abstract
In rats, normobaric hypoxia significantly reduced left ventricular (LV) inotropic function while right ventricular (RV) function was not impaired. In parallel, the animals developed pulmonary edema and inflammation. In the present study, we investigated whether cardiac function and pulmonary injury would aggravate after three and six days of hypoxia exposure or whether cardiopulmonary reactions to prolonged hypoxia would become weaker due to hypoxic acclimatization. Sixty-four female rats were exposed for 72 or 144 h to normoxia. They received a low-rate infusion (0.1 mL/h) with 0.9% NaCl solution. We evaluated indicators of the general condition, blood gas parameters, and hemodynamic function of the rats. In addition, we performed histological and immunohistochemical analyses of the lung. Despite a significant increase in hemoglobin concentration, the LV function deteriorated with prolonged hypoxia. In contrast, the RV systolic pressure and contractility steadily increased by six days of hypoxia. The pulmonary edema and inflammation persisted and rather increased with prolonged hypoxia. Furthermore, elevated protein concentration in the pleural fluid indicated capillary wall stress, which may have aggravated the pulmonary edema. In conclusion, six days of hypoxia and NaCl infusion place significant stress on the cardiopulmonary system of rats, as is also reflected by the 33% of premature deaths in this rat group.
Collapse
Affiliation(s)
- Charly Bambor
- Carl-Ludwig-Institute of Physiology, University of Leipzig, 04103 Leipzig, Germany; (C.B.); (S.D.)
| | - Sarah Daunheimer
- Carl-Ludwig-Institute of Physiology, University of Leipzig, 04103 Leipzig, Germany; (C.B.); (S.D.)
| | - Coralie Raffort
- Department of Pediatric Cardiology, Heart Centre, University of Leipzig, 04289 Leipzig, Germany; (C.R.); (A.S.)
| | - Julia Koedel
- Institute of Pathology, University of Leipzig, 04103 Leipzig, Germany;
| | - Aida Salameh
- Department of Pediatric Cardiology, Heart Centre, University of Leipzig, 04289 Leipzig, Germany; (C.R.); (A.S.)
| | - Beate Raßler
- Carl-Ludwig-Institute of Physiology, University of Leipzig, 04103 Leipzig, Germany; (C.B.); (S.D.)
| |
Collapse
|
16
|
Kostopoulou A, Rebnegger C, Ferrero‐Bordera B, Mattanovich M, Maaß S, Becher D, Gasser B, Mattanovich D. Impact of Oxygen Availability on the Organelle-Specific Redox Potentials and Stress in Recombinant Protein Producing Komagataella phaffii. Microb Biotechnol 2025; 18:e70106. [PMID: 39937160 PMCID: PMC11816699 DOI: 10.1111/1751-7915.70106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/17/2025] [Accepted: 01/24/2025] [Indexed: 02/13/2025] Open
Abstract
The yeast Komagataella phaffii (syn. Pichia pastoris) is a highly effective and well-established host for the production of recombinant proteins. The redox balance of its secretory pathway, which is multi-organelle dependent, is of high importance for producing secretory proteins. Redox imbalance and oxidative stress can significantly influence protein folding and secretion. Glutathione serves as the main redox buffer of the cell and cellular redox conditions can be assessed through the status of the glutathione redox couple (GSH-GSSG). Previous research often focused on the redox potential of the endoplasmic reticulum (ER), where oxidative protein folding and disulphide bond formation occur. In this study, in vivo measurements of the glutathione redox potential were extended to different subcellular compartments by targeting genetically encoded redox sensitive fluorescent proteins (roGFPs) to the cytosol, ER, mitochondria and peroxisomes. Using these biosensors, the impact of oxygen availability on the redox potentials of the different organelles was investigated in non-producing and producing K. phaffii strains in glucose-limited chemostat cultures. It was found that the transition from normoxic to hypoxic conditions affected the redox potential of all investigated organelles, while the exposure to hyperoxic conditions did not impact them. Also, as reported previously, hypoxic conditions led to increased recombinant protein secretion. Finally, transcriptome and proteome analyses provided novel insights into the short-term response of the cells from normoxic to hypoxic conditions.
Collapse
Grants
- Österreichische Forschungsförderungsgesellschaft
- 813979 Horizon 2020 Framework Programme
- Austrian Federal Ministry of Labour and Economy (BMAW), the Austrian Federal Ministry of Climate Action, Environment, Energy, Mobility, Innovation and Technology (BMK), the Styrian Business Promotion Agency SFG, the Standortagentur Tirol, the Government of Lower Austria, the Business Agency Vienna and BOKU through the COMET Funding Program managed by the Austrian Research Promotion Agency FFG, the Nationalstiftung FTE and the Christian Doppler Research Association
- Österreichische Forschungsförderungsgesellschaft
- Horizon 2020 Framework Programme
- Austrian Federal Ministry of Labour and Economy (BMAW), the Austrian Federal Ministry of Climate Action, Environment, Energy, Mobility, Innovation and Technology (BMK), the Styrian Business Promotion Agency SFG, the Standortagentur Tirol, the Government of Lower Austria, the Business Agency Vienna and BOKU through the COMET Funding Program managed by the Austrian Research Promotion Agency FFG, the Nationalstiftung FTE and the Christian Doppler Research Association
Collapse
Affiliation(s)
- Aliki Kostopoulou
- Austrian Centre of Industrial Biotechnology (ACIB)ViennaAustria
- Department of Biotechnology and Food ScienceInstitute of Microbiology and Microbial Biotechnology, BOKU UniversityViennaAustria
| | - Corinna Rebnegger
- Austrian Centre of Industrial Biotechnology (ACIB)ViennaAustria
- Department of Biotechnology and Food ScienceInstitute of Microbiology and Microbial Biotechnology, BOKU UniversityViennaAustria
- Department of Biotechnology and Food Science, Christian Doppler Laboratory for Growth Decoupled Protein Production in YeastBOKU UniversityViennaAustria
| | - Borja Ferrero‐Bordera
- Department of Microbial ProteomicsInstitute of Microbiology, University of GreifswaldGreifswaldGermany
| | - Matthias Mattanovich
- Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Sandra Maaß
- Department of Microbial ProteomicsInstitute of Microbiology, University of GreifswaldGreifswaldGermany
| | - Dörte Becher
- Department of Microbial ProteomicsInstitute of Microbiology, University of GreifswaldGreifswaldGermany
| | - Brigitte Gasser
- Austrian Centre of Industrial Biotechnology (ACIB)ViennaAustria
- Department of Biotechnology and Food ScienceInstitute of Microbiology and Microbial Biotechnology, BOKU UniversityViennaAustria
- Department of Biotechnology and Food Science, Christian Doppler Laboratory for Growth Decoupled Protein Production in YeastBOKU UniversityViennaAustria
| | - Diethard Mattanovich
- Austrian Centre of Industrial Biotechnology (ACIB)ViennaAustria
- Department of Biotechnology and Food ScienceInstitute of Microbiology and Microbial Biotechnology, BOKU UniversityViennaAustria
| |
Collapse
|
17
|
Gorini F, Tonacci A. Ambient Air Pollution and Congenital Heart Disease: Updated Evidence and Future Challenges. Antioxidants (Basel) 2025; 14:48. [PMID: 39857382 PMCID: PMC11761577 DOI: 10.3390/antiox14010048] [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: 12/08/2024] [Revised: 12/28/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Congenital heart disease (CHD) represents the major cause of infant mortality related to congenital anomalies globally. The etiology of CHD is mostly multifactorial, with environmental determinants, including maternal exposure to ambient air pollutants, assumed to contribute to CHD development. While particulate matter (PM) is responsible for millions of premature deaths every year, overall ambient air pollutants (PM, nitrogen and sulfur dioxide, ozone, and carbon monoxide) are known to increase the risk of adverse pregnancy outcomes. In this literature review, we provide an overview regarding the updated evidence related to the association between maternal exposure to outdoor air pollutants and CHD occurrence, also exploring the underlying biological mechanisms from human and experimental studies. With the exception of PM, for which there is currently moderate evidence of its positive association with overall CHD risk following exposure during the periconception and throughout pregnancy, and for ozone which shows a signal of association with increased risk of pooled CHD and certain CHD subtypes in the periconceptional period, for the other pollutants, the data are inconsistent, and no conclusion can be drawn about their role in CHD onset. Future epidemiological cohort studies in countries with different degree of air pollution and experimental research on animal models are warranted to gain a comprehensive picture of the possible involvement of ambient air pollutants in CHD etiopathogenesis. While on the one hand this information could also be useful for timely intervention to reduce the risk of CHD, on the other hand, it is mandatory to scale up the use of technologies for pollutant monitoring, as well as the use of Artificial Intelligence for data analysis to identify the non-linear relationships that will eventually exist between environmental and clinical variables.
Collapse
Affiliation(s)
- Francesca Gorini
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy;
| | | |
Collapse
|
18
|
Li YC, Cheng ML. Carvedilol Confers Ferroptosis Resistance in HL-1 Cells by Upregulating GPX4, FTH1, and FTL1 and Inducing Metabolic Remodeling Under Hypoxia/Reoxygenation. Antioxidants (Basel) 2024; 14:7. [PMID: 39857341 PMCID: PMC11762394 DOI: 10.3390/antiox14010007] [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: 10/11/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Hypoxia/reoxygenation (HR) often occurs under cardiac pathological conditions, and HR-induced oxidative stress usually leads to cardiomyocyte damage. Carvedilol, a non-selective β-blocker, is used clinically to treat cardiac ischemia diseases. Moreover, Carvedilol has also been reported to have an antioxidant ability by reducing lipid peroxidation. However, the mechanism of Carvedilol to inhibit lipid peroxidation is still elusive. To explore the protective mechanism of Carvedilol to resist lipid peroxidation on cardiomyocytes, HL-1 cells were cultured under normoxia, hypoxia, and HR and treated with Carvedilol to investigate the alteration on metabolism, protein expression, and mRNA level to explain its oxidative mechanism. The study found that Carvedilol upregulated glutathione peroxidase 4 (GPX4) protein expression to resist HR-induced lipid peroxidation by metabolic remodeling under HR. Also, Carvedilol promoted ferroptosis-related genes, ferritin heavy chain 1 (FTH1) and ferritin light chain 1 (FTL1) mRNA levels, to reduce lipid peroxidation under both hypoxia and HR. In conclusion, our study explores a mechanism by which Carvedilol inhibits ferroptosis by upregulating GPX4, FTH1, and FTL1 levels to downregulate lipid peroxidation under HR. The study provides a potential strategy for using Carvedilol in clinical applications, inspiring further research and development in the area of heart diseases.
Collapse
Affiliation(s)
- Yi-Chin Li
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan;
| | - Mei-Ling Cheng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan;
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan City 33302, Taiwan
- Clinical Metabolomics Core Laboratory, Chang Gung Memorial Hospital, Taoyuan City 33305, Taiwan
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| |
Collapse
|
19
|
Lv R, Zhao Y, Wang X, He Y, Dong N, Min X, Liu X, Yu Q, Yuan K, Yue H, Yin Q. GLP-1 analogue liraglutide attenuates CIH-induced cognitive deficits by inhibiting oxidative stress, neuroinflammation, and apoptosis via the Nrf2/HO-1 and MAPK/NF-κB signaling pathways. Int Immunopharmacol 2024; 142:113222. [PMID: 39321702 DOI: 10.1016/j.intimp.2024.113222] [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: 07/31/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Obstructive sleep apnea (OSA) is a common clinical condition linked to cognitive impairment, mainly characterized by chronic intermittent hypoxia (CIH). GLP-1 receptor agonist, known for promoting insulin secretion and reducing glucose levels, has demonstrated neuroprotective effects in various experimental models such as stroke, Alzheimer's disease, and Parkinson's disease. This study aims to investigate the potential role and mechanisms of the GLP-1 receptor agonist liraglutide in ameliorating OSA-induced cognitive deficits. CIH exposure, a well-established and mature OSA pathological model, was used both in vitro and in vivo. In vitro, CIH significantly activated oxidative stress, inflammation, and apoptosis in SH-SY5Y cells. Liraglutide enhanced the nuclear translocation of Nrf2, activating its downstream pathways, thereby mitigating CIH-induced injury in SH-SY5Y cells. Additionally, liraglutide modulated the MAPK/NF-κB signaling pathway, reducing the expression of inflammatory factors and proteins. In vivo, we subjected mice to an intermittent hypoxia incubator to mimic the pathogenesis of human OSA. The Morris water maze test revealed that CIH exposure substantially impaired spatial memory. Subsequent western blot analyses and histopathological examinations indicated that liraglutide could activate the Nrf2/HO-1 axis and inhibit the MAPK/NF-κB signaling pathway, thereby alleviating OSA-associated cognitive dysfunction in mice. These findings suggest that GLP-1 receptor agonists may offer a promising preventive strategy for OSA-associated cognitive impairment. By refining these findings, we provide new insights into GLP-1's protective mechanisms in combating cognitive deficits associated with CIH, underscoring its potential as a therapeutic agent for conditions linked to OSA.
Collapse
Affiliation(s)
- Renjun Lv
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Yan Zhao
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Xiao Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Yao He
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Na Dong
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Xiangzhen Min
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Xueying Liu
- Jinan Third People's Hospital, Jinan, Shandong 250132, China
| | - Qin Yu
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Kai Yuan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Chinese Academy of Medical Sciences Research Unit (No. 2018RU006), Peking University, Beijing, China
| | - Hongmei Yue
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| | - Qingqing Yin
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China; Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China.
| |
Collapse
|
20
|
Sandeep, Subba R, Mondal AC. Does COVID-19 Trigger the Risk for the Development of Parkinson's Disease? Therapeutic Potential of Vitamin C. Mol Neurobiol 2024; 61:9945-9960. [PMID: 37957424 DOI: 10.1007/s12035-023-03756-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), which was proclaimed a pandemic by the World Health Organization (WHO) in March 2020. There is mounting evidence that older patients with multimorbidity are more susceptible to COVID-19 complications than are younger, healthy people. Having neuroinvasive potential, SARS-CoV-2 infection may increase susceptibility toward the development of Parkinson's disease (PD), a progressive neurodegenerative disorder with extensive motor deficits. PD is characterized by the aggregation of α-synuclein in the form of Lewy bodies and the loss of dopaminergic neurons in the dorsal striatum and substantia nigra pars compacta (SNpc) of the nigrostriatal pathway in the brain. Increasing reports suggest that SARS-CoV-2 infection is linked with the worsening of motor and non-motor symptoms with high rates of hospitalization and mortality in PD patients. Common pathological changes in both diseases involve oxidative stress, mitochondrial dysfunction, neuroinflammation, and neurodegeneration. COVID-19 exacerbates the damage ensuing from the dysregulation of those processes, furthering neurological complications, and increasing the severity of PD symptomatology. Phytochemicals have antioxidant, anti-inflammatory, and anti-apoptotic properties. Vitamin C supplementation is found to ameliorate the common pathological changes in both diseases to some extent. This review aims to present the available evidence on the association between COVID-19 and PD, and discusses the therapeutic potential of vitamin C for its better management.
Collapse
Affiliation(s)
- Sandeep
- Laboratory of Cellular & Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Rhea Subba
- Laboratory of Cellular & Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Amal Chandra Mondal
- Laboratory of Cellular & Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
21
|
Wang X, Jowsey WJ, Cheung CY, Smart CJ, Klaus HR, Seeto NE, Waller NJ, Chrisp MT, Peterson AL, Ofori-Anyinam B, Strong E, Nijagal B, West NP, Yang JH, Fineran PC, Cook GM, Jackson SA, McNeil MB. Whole genome CRISPRi screening identifies druggable vulnerabilities in an isoniazid resistant strain of Mycobacterium tuberculosis. Nat Commun 2024; 15:9791. [PMID: 39537607 PMCID: PMC11560980 DOI: 10.1038/s41467-024-54072-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Drug-resistant strains of Mycobacterium tuberculosis are a major global health problem. Resistance to the front-line antibiotic isoniazid is often associated with mutations in the katG-encoded bifunctional catalase-peroxidase. We hypothesise that perturbed KatG activity would generate collateral vulnerabilities in isoniazid-resistant katG mutants, providing potential pathway targets to combat isoniazid resistance. Whole genome CRISPRi screens, transcriptomics, and metabolomics were used to generate a genome-wide map of cellular vulnerabilities in an isoniazid-resistant katG mutant strain of M. tuberculosis. Here, we show that metabolic and transcriptional remodelling compensates for the loss of KatG but in doing so generates vulnerabilities in respiration, ribosome biogenesis, and nucleotide and amino acid metabolism. Importantly, these vulnerabilities are more sensitive to inhibition in an isoniazid-resistant katG mutant and translated to clinical isolates. This work highlights how changes in the physiology of drug-resistant strains generates druggable vulnerabilities that can be exploited to improve clinical outcomes.
Collapse
Affiliation(s)
- XinYue Wang
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - William J Jowsey
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Chen-Yi Cheung
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Caitlan J Smart
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Hannah R Klaus
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Noon Ej Seeto
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Natalie Je Waller
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Michael T Chrisp
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Amanda L Peterson
- Metabolomics Australia, Bio21 Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Boatema Ofori-Anyinam
- Center for Emerging and Re-emerging Pathogens, Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Emily Strong
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Brunda Nijagal
- Metabolomics Australia, Bio21 Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Nicholas P West
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Jason H Yang
- Center for Emerging and Re-emerging Pathogens, Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Peter C Fineran
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- Genetics Otago, University of Otago, Dunedin, New Zealand
- Bio-Protection Research Centre, University of Otago, Dunedin, New Zealand
| | - Gregory M Cook
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- School of Biomedical Sciences, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Simon A Jackson
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Matthew B McNeil
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
22
|
Zhang X, Wang X, Yin L, Wang D, Jiao H, Liu X, Zheng J. HACE1 exerts a neuroprotective role against oxidative stress in cerebral ischemia-reperfusion injury by activating the PI3K/AKT/Nrf2 pathway. Neuroscience 2024; 559:249-262. [PMID: 39244008 DOI: 10.1016/j.neuroscience.2024.09.002] [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: 03/22/2024] [Revised: 08/26/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
HECT domain and Ankyrin repeat-containing E3 ubiquitin protein ligase 1 (HACE1) is an E3 ubiquitin ligase involving oxidative stress, an important contributor in cerebral ischemia-reperfusion injury (CIRI). It was proposed to be associated with the PI3K/AKT pathway and Nrf2 nuclear translocation, which are important players of oxidative stress. Therefore, we supposed that HACE1 might affect CIRI by regulating the PI3K/AKT/Nrf2 pathway. Here, we used the transient middle cerebral artery occlusion-reperfusion (tMCAO/R) model to induce CIRI in rats and found lower HACE1 expression in ischemic rats compared with the control. To explore the exact role of HACE1, the lentivirus vector carrying the HACE1 sequence was administrated to rats by intracerebroventricular injection (1 × 109 TU/mL, 9 μL) one week before tMCAO/R operation. HACE1 overexpression alleviated tMCAO/R-induced brain damage in rats. Further studies revealed that it reduced oxidative stress via activating the PI3K/AKT/Nrf2 pathway, thereby inhibiting neuronal apoptosis in the ischemic penumbra of rats with CIRI. Then, differentiated PC12 cells were cultured in oxygen-glucose deprivation-reoxygenation (OGD/R) conditions (OGD: 1 % O2, 94 % N2, and 5 % CO2; R: normal atmosphere) to simulate CIRI in vitro. Similarly, HACE1 overexpression inhibited neuronal apoptosis caused by OGD/R treatment. The PI3K inhibitor LY294002 reversed the inhibitory effects of HACE1 overexpression on oxidative stress in OGD/R-injured cells, accompanied by the inactivated AKT/Nrf2 pathway. Altogether, our results suggest that HACE1 protects against oxidative stress-induced neuronal apoptosis in CIRI by activating the PI3K/AKT/Nrf2 pathway, providing a new insight into the CIRI treatment.
Collapse
Affiliation(s)
- Xinyue Zhang
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Xiao Wang
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Le Yin
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Dan Wang
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Hong Jiao
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Xiaodan Liu
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Jiaolin Zheng
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China.
| |
Collapse
|
23
|
Alves JL, Reis PM, Quinta-Ferreira RM, Quinta-Ferreira ME, Matias CM. Changes in reactive oxygen species and autofluorescence under hypoxia at the hippocampal CA3 area: Role of calcium and zinc influxes. Neurochem Int 2024; 180:105882. [PMID: 39413928 DOI: 10.1016/j.neuint.2024.105882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 09/12/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024]
Abstract
Reactive oxygen species (ROS) have an important role in cellular biology, being involved, in a way that depends on their levels, in cell signaling processes or in oxidative stress, probably associated with neurodegenerative and other diseases. Most of the studies about ROS formation were performed in ischemic conditions, and thus, there is limited knowledge about ROS formation in less severe hypoxic conditions. This study investigates neuronal ROS generation and autofluorescence changes in hypoxic conditions, focusing on the involvement of calcium and zinc. Using hippocampal slices from Wistar rats, ROS production was monitored by the permeant fluorescent indicator H2DCFDA under different oxygenation levels. Moderate hypoxia (40% O2) led to a small ROS increase, while severe hypoxia (0% O2) showed a more pronounced rise. KCl-induced depolarization significantly enhanced ROS formation, particularly under severe hypoxia. Inhibition of NMDA receptors reduced ROS generation without affecting autofluorescence, while chelation of zinc ions decreased ROS production and increased flavin adenine dinucleotide (FAD) autofluorescence. These findings suggest that, in hypoxic conditions, ROS formation is mediated by calcium entry through NMDA receptors and also by zinc influxes. Thus, these ions play a crucial role in oxidative stress, which may be related with neurodegenerative diseases associated with ROS dysregulation.
Collapse
Affiliation(s)
- João L Alves
- Department of Life Sciences, University of Coimbra, Portugal; CNC-UC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal
| | - Patrícia M Reis
- CERES - Chemical Engineering and Renewable Resources for Sustainability, Department of Chemical Engineering, University of Coimbra, Portugal
| | - Rosa M Quinta-Ferreira
- CERES - Chemical Engineering and Renewable Resources for Sustainability, Department of Chemical Engineering, University of Coimbra, Portugal
| | - M Emília Quinta-Ferreira
- CNC-UC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal; Department of Physics, University of Coimbra, Portugal
| | - Carlos M Matias
- CNC-UC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal; Department of Physics, UTAD, Vila Real, Portugal.
| |
Collapse
|
24
|
Wang X, Fan F, Hou Y, Meng X. Tile: Construction of a specific nanoprobe for scavenging ROS in hypobaric hypoxia induced brain injury of mice. Heliyon 2024; 10:e38958. [PMID: 39640698 PMCID: PMC11620081 DOI: 10.1016/j.heliyon.2024.e38958] [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: 10/30/2023] [Revised: 09/28/2024] [Accepted: 10/03/2024] [Indexed: 12/07/2024] Open
Abstract
The prevention and treatment of hypobaric hypoxia brain injury (HHBI) remains an unprecedented challenge due to the complex oxidative stress response at the damage site. In this study, RuCO phthalocyanine compound (RuPc) and bovine serum albumin (BSA) were self-assembled to obtain RuPc-BSA nanoparticles for HHBI therapy. As a nanoprobe carrying and storing carbon monoxide (CO), RuPc-BSA delivers CO to pathologically damaged areas of the brain. CO specifically attaches itself to the heme functional groups on mitochondria and restricts the source of reactive oxygen species (ROS) generation. RuPc-BSA nanoparticles have been demonstrated in vitro to exhibit amazing stability as well as remarkable scavenging activity on hydroxyl radical, superoxide anion, and hydrogen peroxide. In vivo experiments showed that ROS levels in the brain of HHBI rats pretreated with RuPc-BSA decreased significantly, and neuronal function and oxidative stress levels were alleviated. Western blot and qRT-RCR results indicated that RuPc-BSA restricted the protein levels of Keap1, whereas enhanced the gene and protein levels of Nrf2. This study demonstrated that RuPc-BSA can ameliorate HHBI of mice by scavenging ROS partly via activating Keap1/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Xiaobo Wang
- Innovative Institute of Chinese Medicine and Pharmacy/Academy for Interdiscipline, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Fuhan Fan
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ya Hou
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
| | - Xianli Meng
- Innovative Institute of Chinese Medicine and Pharmacy/Academy for Interdiscipline, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
| |
Collapse
|
25
|
Li J, Yang D, Lin L, Yu L, Chen L, Lu K, Lan J, Zeng Y, Xu Y. Important functions and molecular mechanisms of aquaporins family on respiratory diseases: potential translational values. J Cancer 2024; 15:6073-6085. [PMID: 39440058 PMCID: PMC11493008 DOI: 10.7150/jca.98829] [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: 05/24/2024] [Accepted: 08/25/2024] [Indexed: 10/25/2024] Open
Abstract
Aquaporins (AQPs) are a subgroup of small transmembrane transporters that are distributed in various types of tissues, including the lung, kidney, heart and central nervous system. It is evident that respiratory diseases represent a significant global health concern, with a considerable number of deaths occurring worldwide. Recent researches have demonstrated that AQPs play a pivotal role in respiratory diseases, including chronic obstructive pulmonary disease (COPD), asthma, acute respiratory distress syndrome (ARDS), and particularly non-small cell lung cancer (NSCLC). In the context of NSCLC, the overexpression of AQP1, AQP3, AQP4, and AQP5 has been demonstrated to facilitate tumor angiogenesis, as well as the proliferation, migration, and invasiveness of tumor cells. This review concisely explores the role of AQP family on respiratory diseases, to assess their clinical and translational significance for understanding molecular pathogenesis. However, the potential translation of AQPs biomarkers into clinical applications is promising and the understanding of the precise mechanisms influencing respiratory diseases is still ongoing. Addressing the challenges and outlining the future perspectives in AQPs development is essential for clinical progress in a concise manner.
Collapse
Affiliation(s)
- Jinshan Li
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Clinical Research Center of Interventional Respirology, Quanzhou, Fujian Province, 362000, China
| | - Dongyong Yang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
| | - Lanlan Lin
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Clinical Research Center of Interventional Respirology, Quanzhou, Fujian Province, 362000, China
| | - Liying Yu
- Central Laboratory, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
| | - Luyang Chen
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Clinical Research Center of Interventional Respirology, Quanzhou, Fujian Province, 362000, China
| | - Kaiqiang Lu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Clinical Research Center of Interventional Respirology, Quanzhou, Fujian Province, 362000, China
| | - Jieli Lan
- Clinical Research Unit, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Yiming Zeng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Clinical Research Center of Interventional Respirology, Quanzhou, Fujian Province, 362000, China
| | - Yuan Xu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Clinical Research Center of Interventional Respirology, Quanzhou, Fujian Province, 362000, China
- School of Public Health, Fujian Medical University, Fuzhou, Fujian Province, 350000, China
| |
Collapse
|
26
|
Sahoo AK, Chivukula N, Madgaonkar SR, Ramesh K, Marigoudar SR, Sharma KV, Samal A. Leveraging integrative toxicogenomic approach towards development of stressor-centric adverse outcome pathway networks for plastic additives. Arch Toxicol 2024; 98:3299-3321. [PMID: 39097536 PMCID: PMC11402864 DOI: 10.1007/s00204-024-03825-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/24/2024] [Indexed: 08/05/2024]
Abstract
Plastics are widespread pollutants found in atmospheric, terrestrial and aquatic ecosystems due to their extensive usage and environmental persistence. Plastic additives, that are intentionally added to achieve specific functionality in plastics, leach into the environment upon plastic degradation and pose considerable risk to ecological and human health. Limited knowledge concerning the presence of plastic additives throughout plastic life cycle has hindered their effective regulation, thereby posing risks to product safety. In this study, we leveraged the adverse outcome pathway (AOP) framework to understand the mechanisms underlying plastic additives-induced toxicities. We first identified an exhaustive list of 6470 plastic additives from chemicals documented in plastics. Next, we leveraged heterogenous toxicogenomics and biological endpoints data from five exposome-relevant resources, and identified associations between 1287 plastic additives and 322 complete and high quality AOPs within AOP-Wiki. Based on these plastic additive-AOP associations, we constructed a stressor-centric AOP network, wherein the stressors are categorized into ten priority use sectors and AOPs are linked to 27 disease categories. We visualized the plastic additives-AOP network for each of the 1287 plastic additives and made them available in a dedicated website: https://cb.imsc.res.in/saopadditives/ . Finally, we showed the utility of the constructed plastic additives-AOP network by identifying highly relevant AOPs associated with benzo[a]pyrene (B[a]P), bisphenol A (BPA), and bis(2-ethylhexyl) phthalate (DEHP) and thereafter, explored the associated toxicity pathways in humans and aquatic species. Overall, the constructed plastic additives-AOP network will assist regulatory risk assessment of plastic additives, thereby contributing towards a toxic-free circular economy for plastics.
Collapse
Affiliation(s)
- Ajaya Kumar Sahoo
- Computational Biology Group, The Institute of Mathematical Sciences (IMSc), CIT Campus, Taramani, Chennai, 600113, India
- Homi Bhabha National Institute (HBNI), Mumbai, 400094, India
| | - Nikhil Chivukula
- Computational Biology Group, The Institute of Mathematical Sciences (IMSc), CIT Campus, Taramani, Chennai, 600113, India
- Homi Bhabha National Institute (HBNI), Mumbai, 400094, India
| | - Shreyes Rajan Madgaonkar
- Computational Biology Group, The Institute of Mathematical Sciences (IMSc), CIT Campus, Taramani, Chennai, 600113, India
- Homi Bhabha National Institute (HBNI), Mumbai, 400094, India
| | - Kundhanathan Ramesh
- Computational Biology Group, The Institute of Mathematical Sciences (IMSc), CIT Campus, Taramani, Chennai, 600113, India
| | | | - Krishna Venkatarama Sharma
- Ministry of Earth Sciences, National Centre for Coastal Research, Government of India, Pallikaranai, Chennai, 600100, India
| | - Areejit Samal
- Computational Biology Group, The Institute of Mathematical Sciences (IMSc), CIT Campus, Taramani, Chennai, 600113, India.
- Homi Bhabha National Institute (HBNI), Mumbai, 400094, India.
| |
Collapse
|
27
|
Gokdemir GS, Seker U, Demirtas B, Taskin S. Effects of acute carbon monoxide poisoning on liver damage and comparisons of related oxygen therapies in a rat model. Toxicol Mech Methods 2024; 34:845-854. [PMID: 38721843 DOI: 10.1080/15376516.2024.2353887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/20/2024] [Accepted: 05/04/2024] [Indexed: 05/24/2024]
Abstract
Acute carbon monoxide (CO) poisoning may cause liver damage and liver dysfunction. Therefore, in this study, we aimed to compare the efficiency of normobaric oxygen (NBO) and high-flow nasal cannula oxygen (HFNCO) treatments on liver injury. For that purpose, 28 male Wistar albino rats were divided into four groups (Control, CO, CO + NBO, and CO + HFNCO). The control group was allowed to breath room air for 30 min. Acute CO poisoning in CO, CO + NBO, CO + HFNCO was induced by CO exposure for 30 min. Thereafter, NBO group received 100% NBO with reservoir mask for 30 min. HFNCO group received high-flow oxygen through nasal cannula for 30 min. At the end of the experiment, all animals were sacrificed by cardiac puncture under anesthesia. Serum liver function tests were measured. Liver tissue total antioxidant status (TAS), total oxidant status (TOS), and oxidative stress index (OSI) levels, tissue histomorphology and immunoexpression levels of Bax, Caspase 3, TNF-α, IL-1β, and NF-κB were also examined. Our observations indicated that acute CO poisoning caused significant increases in blood COHb, serum aminotransferase (AST), alanine aminotransferase (ALT0, alkaline phosphatase (ALP), total protein, albumin, and globulin levels but a decrease in albumin to globulin ratio (all, p < 0.05). Furthermore, acute CO poisoning significantly increased the OSI value, and the immunoexpresssion of Bax, Caspase 3, TNF-α, IL-1β, and NF-κB in liver tissue (all, p < 0.05). These pathological changes in serum and liver tissue were alleviated through both of the treatment methods. In conclusion, both the NBO and HFNCO treatments were beneficial to alleviate the acute CO poisoning associated with liver injury and dysfunction.
Collapse
Affiliation(s)
- Gul Sahika Gokdemir
- Department of Physiology, Faculty of Medicine, Mardin Artuklu University, Mardin, Turkey
| | - Ugur Seker
- Department of Histology and Embryology, Faculty of Medicine, Mardin Artuklu University, Mardin, Turkey
| | - Berjan Demirtas
- Vocational School Veterinary Medicine, Plant and Animal Production, Equine and Training Program, İstanbul University-Cerrahpaşa, İstanbul, Turkey
| | - Seyhan Taskin
- Department of Physiology, Faculty of Medicine, Harran University, Sanliurfa, Turkey
| |
Collapse
|
28
|
Malovic E, Ealy A, Miller C, Jang A, Hsu PJ, Sarkar S, Rokad D, Goeser C, Hartman AK, Zhu A, Palanisamy B, Zenitsky G, Jin H, Anantharam V, Kanthasamy A, He C, Kanthasamy AG. Epitranscriptomic reader YTHDF2 regulates SEK1( MAP2K4)-JNK-cJUN inflammatory signaling in astrocytes during neurotoxic stress. iScience 2024; 27:110619. [PMID: 39252959 PMCID: PMC11382029 DOI: 10.1016/j.isci.2024.110619] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 06/13/2024] [Accepted: 07/26/2024] [Indexed: 09/11/2024] Open
Abstract
As the most abundant glial cells in the central nervous system (CNS), astrocytes dynamically respond to neurotoxic stress, however, the key molecular regulators controlling the inflammatory status of these sentinels during neurotoxic stress are many and complex. Herein, we demonstrate that the m6A epitranscriptomic mRNA modification tightly regulates the pro-inflammatory functions of astrocytes. Specifically, the astrocytic neurotoxic stressor, manganese (Mn), downregulated the m6A reader YTHDF2 in human and mouse astrocyte cultures and in the mouse brain. Functionally, YTHDF2 knockdown augmented, while its overexpression dampened, the neurotoxic stress-induced proinflammatory response, suggesting YTHDF2 serves as a key upstream regulator of inflammatory responses in astrocytes. Mechanistically, YTHDF2 RIP-sequencing identified MAP2K4 (MKK4; SEK1) mRNA as a YTHDF2 target influencing inflammatory signaling. Our target validation revealed that Mn-exposed astrocytes mediate proinflammatory responses by activating the phosphorylation of SEK1, JNK, and cJUN signaling. Collectively, YTHDF2 serves as a key upstream 'molecular switch' controlling SEK1(MAP2K4)-JNK-cJUN proinflammatory signaling in astrocytes.
Collapse
Affiliation(s)
- Emir Malovic
- Parkinson's Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Alyssa Ealy
- Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, USA
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Cameron Miller
- Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, USA
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Ahyoung Jang
- Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, USA
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Phillip J Hsu
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Souvarish Sarkar
- Parkinson's Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Dharmin Rokad
- Parkinson's Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Cody Goeser
- Parkinson's Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Aleah Kristen Hartman
- Parkinson's Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Allen Zhu
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Bharathi Palanisamy
- Parkinson's Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Gary Zenitsky
- Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, USA
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Huajun Jin
- Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, USA
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Vellareddy Anantharam
- Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, USA
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Arthi Kanthasamy
- Parkinson's Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
- Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, USA
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Chuan He
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Anumantha G Kanthasamy
- Parkinson's Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
- Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, USA
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| |
Collapse
|
29
|
Tayman C, Çakır U, Kurt A, Ertekin Ö, Taskin Turkmenoglu T, Çağlayan M, Işık E. Evaluation of beneficial effects of dexpanthenol on hypoxic-ischemic encephalopathy. Biotech Histochem 2024; 99:260-268. [PMID: 38869860 DOI: 10.1080/10520295.2024.2365231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a cause of serious morbidity and mortality in newborns. Dexpanthenol, which is metabolized into D-pantothenic acid, has antioxidant and other potentially therapeutic properties. We examined some effects of dexpanthenol on the brains of week-old rat pups with HIE induced by obstruction of the right carotid artery followed by keeping in 8% O2 for 2 hours. Dexpanthenol (500 mg/kg) was administered intraperitoneally to 16 of 32 pups with HIE. Protein, DNA, and lipid oxidation degradation products were assayed and hippocampal and cortical cell apoptosis and neuronal cell numbers were evaluated in stained sections. Dexpanthenol application reduced oxidative stress and inflammation. TNF-α and IL-6 cytokine levels in HIE also decreased with dexpanthenol treatment. The numbers of caspase-3 positive cells in the dentate gyrus and CA1/CA2/CA3 regions of the hippocampus was lower, and apoptosis was decreased in the dexpanthenol-treated animals. These findings suggest possible clinical applications of dexpanthenol in human HIE.
Collapse
Affiliation(s)
- Cuneyt Tayman
- Department of Neonatology, SBU Ankara City Hospital, Ankara, Turkey
| | - Ufuk Çakır
- Department of Neonatology, SBU Ankara City Hospital, Ankara, Turkey
| | - Abdullah Kurt
- Department of Neonatology, SBU Ankara City Hospital, Ankara, Turkey
| | - Ömer Ertekin
- Department of Neonatology, SBU Ankara City Hospital, Ankara, Turkey
| | - Tugba Taskin Turkmenoglu
- Department of Pathology, Ankara Dişkapi Yildirim Beyzat Training and Research Hospital, Ankara, Turkey
| | - Murat Çağlayan
- Department of Medical Biochemistry, Dışkapı Yıldırım Beyazıt Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Eray Işık
- Department of Otorhinolaryngology (Ear-Nose-Throat), Ankara Dişkapi Yildirim Beyzat Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
30
|
Gao J, Guo Z, Zhao M, Cheng X, Jiang X, Liu Y, Zhang W, Yue X, Fei X, Jiang Y, Chen L, Zhang S, Zhao T, Zhu L. Lipidomics and mass spectrometry imaging unveil alterations in mice hippocampus lipid composition exposed to hypoxia. J Lipid Res 2024; 65:100575. [PMID: 38866327 PMCID: PMC11333011 DOI: 10.1016/j.jlr.2024.100575] [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: 03/21/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/14/2024] Open
Abstract
Lipids are components of cytomembranes that are involved in various biochemical processes. High-altitude hypoxic environments not only affect the body's energy metabolism, but these environments can also cause abnormal lipid metabolism involved in the hypoxia-induced cognitive impairment. Thus, comprehensive lipidomic profiling of the brain tissue is an essential step toward understanding the mechanism of cognitive impairment induced by hypoxic exposure. In the present study, mice showed reduced new-object recognition and spatial memory when exposed to hypobaric hypoxia for 1 day. Histomorphological staining revealed significant morphological and structural damage to the hippocampal tissue, along with prolonged exposure to hypobaric hypoxia. Dynamic lipidomics of the mouse hippocampus showed a significant shift in both the type and distribution of phospholipids, as verified by spatial lipid mapping. Collectively, a diverse and dynamic lipid composition in mice hippocampus was uncovered, which deepens our understanding of biochemical changes during sustained hypoxic exposure and could provide new insights into the cognitive decline induced by high-altitude hypoxia exposure.
Collapse
Affiliation(s)
- Jiayue Gao
- Department of Brain Plasticity, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Zhiying Guo
- Hepato-pancreato-biliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Ming Zhao
- Department of Brain Plasticity, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Xiang Cheng
- Department of Brain Plasticity, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Xiufang Jiang
- Department of Brain Plasticity, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yikun Liu
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing, China
| | - Wenpeng Zhang
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing, China
| | - Xiangpei Yue
- Department of Brain Plasticity, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Xuechao Fei
- Department of Brain Plasticity, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yaqun Jiang
- Department of Brain Plasticity, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Lu Chen
- Department of Brain Plasticity, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Shaojie Zhang
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Tong Zhao
- Department of Brain Plasticity, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Lingling Zhu
- Department of Brain Plasticity, Beijing Institute of Basic Medical Sciences, Beijing, China; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China.
| |
Collapse
|
31
|
Zhang R, Liu C, Yu D, She D, Yu Y, Cai Y, Chen N. Melatonin protects oogenesis from hypobaric hypoxia-induced fertility damage in mice. ZYGOTE 2024; 32:161-169. [PMID: 38465746 DOI: 10.1017/s0967199424000017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Environmental hypoxia adversely affects reproductive health in humans and animals at high altitudes. Therefore, how to alleviate the follicle development disorder caused by hypoxia exposure and to improve the competence of fertility in plateau non-habituated female animals are important problems to be solved urgently. In this study, a hypobaric hypoxic chamber was used for 4 weeks to simulate hypoxic conditions in female mice, and the effects of hypoxia on follicle development, proliferation and apoptosis of granulosa cells, reactive oxygen species (ROS) levels in MII oocyte and 2-cell rate were evaluated. At the same time, the alleviating effect of melatonin on hypoxic exposure-induced oogenesis damage was evaluated by feeding appropriate amounts of melatonin daily under hypoxia for 4 weeks. The results showed that hypoxia exposure significantly increased the proportion of antral follicles in the ovary, the number of proliferation and apoptosis granulosa cells in the follicle, and the level of ROS in MII oocytes, eventually led to the decline of oocyte quality. However, these defects were alleviated when melatonin was fed under hypoxia conditions. Together, these findings suggest that hypoxia exposure impaired follicular development and reduced oocyte quality, and that melatonin supplementation alleviated the fertility reduction induced by hypoxia exposure.
Collapse
Affiliation(s)
- Ruina Zhang
- School of Biological and Pharmaceutical Engineering West Anhui University, Lu'an, 237012, China
- Anhui Engineering Laboratory for Conservation and Sustainable Utilization of Traditional Chinese Medicine Resources, West Anhui University, Lu'an, 237012, China
| | - Cong Liu
- Center for Reproductive Medicine Renmin Hospital of Wuhan University, Wuhan, 430000, China
| | - Daolun Yu
- School of Biological and Pharmaceutical Engineering West Anhui University, Lu'an, 237012, China
| | - Deyong She
- School of Biological and Pharmaceutical Engineering West Anhui University, Lu'an, 237012, China
| | - Yan Yu
- School of Biological and Pharmaceutical Engineering West Anhui University, Lu'an, 237012, China
| | - Yongping Cai
- College of Life Science, Anhui Agricultural University, Hefei, 230000, China
| | - Naifu Chen
- School of Biological and Pharmaceutical Engineering West Anhui University, Lu'an, 237012, China
- Anhui Engineering Laboratory for Conservation and Sustainable Utilization of Traditional Chinese Medicine Resources, West Anhui University, Lu'an, 237012, China
| |
Collapse
|
32
|
Amro Z, Collins-Praino L, Yool A. Protective roles of peroxiporins AQP0 and AQP11 in human astrocyte and neuronal cell lines in response to oxidative and inflammatory stressors. Biosci Rep 2024; 44:BSR20231725. [PMID: 38451099 PMCID: PMC10965398 DOI: 10.1042/bsr20231725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 03/03/2024] [Accepted: 03/06/2024] [Indexed: 03/08/2024] Open
Abstract
In addition to aquaporin (AQP) classes AQP1, AQP4 and AQP9 known to be expressed in mammalian brain, our recent transcriptomic analyses identified AQP0 and AQP11 in human cortex and hippocampus at levels correlated with age and Alzheimer's disease (AD) status; however, protein localization remained unknown. Roles of AQP0 and AQP11 in transporting hydrogen peroxide (H2O2) in lens and kidney prompted our hypothesis that up-regulation in brain might similarly be protective. Established cell lines for astroglia (1321N1) and neurons (SHSY5Y, differentiated with retinoic acid) were used to monitor changes in transcript levels for human AQPs (AQP0 to AQP12) in response to inflammation (simulated with 10-100 ng/ml lipopolysaccharide [LPS], 24 h), and hypoxia (5 min N2, followed by 0 to 24 h normoxia). AQP transcripts up-regulated in both 1321N1 and SHSY5Y included AQP0, AQP1 and AQP11. Immunocytochemistry in 1321N1 cells confirmed protein expression for AQP0 and AQP11 in plasma membrane and endoplasmic reticulum; AQP11 increased 10-fold after LPS and AQP0 increased 0.3-fold. In SHSY5Y cells, AQP0 expression increased 0.2-fold after 24 h LPS; AQP11 showed no appreciable change. Proposed peroxiporin roles were tested using melondialdehyde (MDA) assays to quantify lipid peroxidation levels after brief H2O2. Boosting peroxiporin expression by LPS pretreatment lowered subsequent H2O2-induced MDA responses (∼50%) compared with controls; conversely small interfering RNA knockdown of AQP0 in 1321N1 increased lipid peroxidation (∼17%) after H2O2, with a similar trend for AQP11 siRNA. Interventions that increase native brain peroxiporin activity are promising as new approaches to mitigate damage caused by aging and neurodegeneration.
Collapse
Affiliation(s)
- Zein Amro
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
| | | | - Andrea J. Yool
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
33
|
Tregub PP, Kulikov VP, Ibrahimli I, Tregub OF, Volodkin AV, Ignatyuk MA, Kostin AA, Atiakshin DA. Molecular Mechanisms of Neuroprotection after the Intermittent Exposures of Hypercapnic Hypoxia. Int J Mol Sci 2024; 25:3665. [PMID: 38612476 PMCID: PMC11011936 DOI: 10.3390/ijms25073665] [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: 02/19/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
The review introduces the stages of formation and experimental confirmation of the hypothesis regarding the mutual potentiation of neuroprotective effects of hypoxia and hypercapnia during their combined influence (hypercapnic hypoxia). The main focus is on the mechanisms and signaling pathways involved in the formation of ischemic tolerance in the brain during intermittent hypercapnic hypoxia. Importantly, the combined effect of hypoxia and hypercapnia exerts a more pronounced neuroprotective effect compared to their separate application. Some signaling systems are associated with the predominance of the hypoxic stimulus (HIF-1α, A1 receptors), while others (NF-κB, antioxidant activity, inhibition of apoptosis, maintenance of selective blood-brain barrier permeability) are mainly modulated by hypercapnia. Most of the molecular and cellular mechanisms involved in the formation of brain tolerance to ischemia are due to the contribution of both excess carbon dioxide and oxygen deficiency (ATP-dependent potassium channels, chaperones, endoplasmic reticulum stress, mitochondrial metabolism reprogramming). Overall, experimental studies indicate the dominance of hypercapnia in the neuroprotective effect of its combined action with hypoxia. Recent clinical studies have demonstrated the effectiveness of hypercapnic-hypoxic training in the treatment of childhood cerebral palsy and diabetic polyneuropathy in children. Combining hypercapnic hypoxia with pharmacological modulators of neuro/cardio/cytoprotection signaling pathways is likely to be promising for translating experimental research into clinical medicine.
Collapse
Affiliation(s)
- Pavel P. Tregub
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- Brain Science Institute, Research Center of Neurology, 125367 Moscow, Russia
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, RUDN University, 117198 Moscow, Russia; (A.V.V.); (M.A.I.); (A.A.K.); (D.A.A.)
| | - Vladimir P. Kulikov
- Department of Ultrasound and Functional Diagnostics, Altay State Medical University, 656040 Barnaul, Russia;
| | - Irada Ibrahimli
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | | | - Artem V. Volodkin
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, RUDN University, 117198 Moscow, Russia; (A.V.V.); (M.A.I.); (A.A.K.); (D.A.A.)
| | - Michael A. Ignatyuk
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, RUDN University, 117198 Moscow, Russia; (A.V.V.); (M.A.I.); (A.A.K.); (D.A.A.)
| | - Andrey A. Kostin
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, RUDN University, 117198 Moscow, Russia; (A.V.V.); (M.A.I.); (A.A.K.); (D.A.A.)
| | - Dmitrii A. Atiakshin
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, RUDN University, 117198 Moscow, Russia; (A.V.V.); (M.A.I.); (A.A.K.); (D.A.A.)
| |
Collapse
|
34
|
Fussing F, Christensson J, Wörtwein G. Expression of erythropoietin receptor protein in the mouse hippocampus in response to normobaric hypoxia. Heliyon 2024; 10:e25051. [PMID: 38322970 PMCID: PMC10844123 DOI: 10.1016/j.heliyon.2024.e25051] [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: 05/07/2023] [Revised: 12/17/2023] [Accepted: 01/18/2024] [Indexed: 02/08/2024] Open
Abstract
Background Over the past decades, accumulating research on erythropoietin (EPO) and its receptor (EPOR) has revealed various neuroprotective actions and upregulation in hypoxic conditions. To our knowledge, EPOR protein levels in the hippocampus and isocortex have never been measured. Therefore, the aim of this study was to measure EPOR protein in the hippocampus (HPC) and prefrontal cortex (PFC). Further objectives were to examine the effects of exposure to normobaric hypoxia of various degrees and durations on EPOR protein and to explore how long-lasting these effects were. Method Adult C57BL/6 mice were randomized into a control group (N = 12) or various hypoxia groups (N = 5-11). Mice were exposed to three different O2 concentrations (10 %, 12 %, or 18 %) for 8 h a day for 5 days and sacrificed immediately after the last exposure. The effect of exposure to 12 % O2 for 1 day and 4 weeks (8 h per day) at this survival time was also examined. Additionally, groups of mice were exposed to 12 % O2 for 1 or 5 days (8 h per day) and euthanized at various times (up to 3 weeks) thereafter to examine the duration of EPOR protein regulation in the HPC and the PFC. EPOR protein was detected with a sandwich-ELISA method. Results EPOR protein was present in the HPC and PFC, at 206.64 ± 43.98 pg/mg and 184.25 ± 48.21 pg/mg, respectively. The highest increase in EPOR protein was observed in the HPC after 5 days of 8 h exposure to 12 % O2 and was most pronounced 24 h after last exposure. The effect of hypoxia normalized within one week after the last exposure. Conclusion This study successfully measured hippocampal EPOR protein and showed a significant association between normobaric hypoxia and acute EPOR elevation. It is our hope that this study can provide guidance to future research on the neuroprotective effects of EPO.
Collapse
Affiliation(s)
- F. Fussing
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen, Mental Health Services, Capital Region of Copenhagen and University Hospital of Copenhagen, Denmark
| | - J. Christensson
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen, Mental Health Services, Capital Region of Copenhagen and University Hospital of Copenhagen, Denmark
| | - G. Wörtwein
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen, Mental Health Services, Capital Region of Copenhagen and University Hospital of Copenhagen, Denmark
- Section of Environmental Health, Department of Public Health, University of Copenhagen, Øster Farimagsgade 5, DK-1014, Copenhagen, Denmark
| |
Collapse
|
35
|
Geng J, Feng J, Ke F, Fang F, Jing X, Tang J, Fang C, Zhang B. MicroRNA-124 negatively regulates STAT3 to alleviate hypoxic-ischemic brain damage by inhibiting oxidative stress. Aging (Albany NY) 2024; 16:2828-2847. [PMID: 38319722 PMCID: PMC10911356 DOI: 10.18632/aging.205513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/29/2023] [Indexed: 02/07/2024]
Abstract
MicroRNA-124 (miR-124) is implicated in various neurological diseases; however, its significance in hypoxic-ischaemic brain damage (HIBD) remains unclear. This study aimed to elucidate the underlying pathophysiological mechanisms of miR-124 in HIBD. In our study performed on oxygen-glucose deprivation followed by reperfusion (OGD)/R-induced primary cortical neurons, a substantial reduction in miR-124 was observed. Furthermore, the upregulation of miR-124 significantly mitigated oxidative stress, apoptosis, and mitochondrial impairment. We demonstrated that miR-124 interacts with the signal transducer and activator of transcription 3 (STAT3) to exert its biological function using the dual-luciferase reporter gene assay. As the duration of OGD increased, miR-124 exhibited a negative correlation with STAT3. STAT3 overexpression notably attenuated the protective effects of miR-124 mimics, while knockdown of STAT3 reversed the adverse effects of the miR-124 inhibitor. Subsequently, we conducted an HIBD model in rats. In vivo experiments, miR-124 overexpression attenuated cerebral infarction volume, cerebral edema, apoptosis, oxidative stress, and improved neurological function recovery in HIBD rats. In summary, the neuroprotective effects of the miR-124/STAT3 axis were confirmed in the HIBD model. MiR-124 may serve as a potential biomarker with significant therapeutic implications for HIBD.
Collapse
Affiliation(s)
- Jiaqing Geng
- Departments of Neonatology, Renmin Hospital of Wuhan University, Wuhan 430062, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430062, China
| | - Jiangpeng Feng
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan 430062, China
| | - Fangzi Ke
- Departments of Neonatology, Renmin Hospital of Wuhan University, Wuhan 430062, China
| | - Fang Fang
- Departments of Neonatology, Renmin Hospital of Wuhan University, Wuhan 430062, China
| | - Xiaoqi Jing
- Departments of Neonatology, Renmin Hospital of Wuhan University, Wuhan 430062, China
| | - Jiaxin Tang
- Departments of Neonatology, Renmin Hospital of Wuhan University, Wuhan 430062, China
| | - Chengzhi Fang
- Departments of Neonatology, Renmin Hospital of Wuhan University, Wuhan 430062, China
| | - Binghong Zhang
- Departments of Neonatology, Renmin Hospital of Wuhan University, Wuhan 430062, China
| |
Collapse
|
36
|
Ma Q, Xu H, Wei Y, Liang M. Effects of acute hypoxia on nutrient metabolism and physiological function in turbot, Scophthalmus maximus. FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:367-383. [PMID: 36609890 DOI: 10.1007/s10695-022-01154-5] [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: 07/05/2022] [Accepted: 11/27/2022] [Indexed: 06/17/2023]
Abstract
Acute hypoxia is a common stress in aquaculture, and causes energy deficiency, oxidative damage and death in fish. Many studies have confirmed that acute hypoxia activated hif1α expression, anaerobic glycolysis and antioxidant system in fish, but the effects of acute hypoxia on lipid and protein metabolism, organelle damage, and the functions of hif2α and hif3α in economic fishes have not been well evaluated. In the present study, turbot was exposed to acute hypoxia (2.0 ± 0.5 mg/L) for 6 h, 12 h, and 24 h, respectively. Then, the contents of hemoglobin (HB), metabolite, gene expressions of hifα isoforms, energy homeostasis, endoplasmic reticulum (ER) stress, and apoptosis were measured. The results suggested that turbot is intolerant to acute hypoxia and the asphyxiation point is about 1.5 mg/L. Acute hypoxia induced perk-mediated ER stress, and increased lipid peroxidation and liver injury in turbot. The blood HB level and liver vegfab expression were increased under hypoxia, which enhances oxygen transport. At hypoxia stress, hif3α, anaerobic glycolysis-related genes expression, and lactate content were increased in the liver, and glycogen was broken down to ensure ATP supply. Meanwhile, hif2α, lipid synthesis-related genes expression, and TG content were increased in the liver, but the lipid catabolism and protein synthesis were suppressed during hypoxia, which reduced the oxygen consumption and ROS generation. Our results systematically illustrate the metabolic and physiological changes under acute hypoxia in turbot, and provide important guidance to improve hypoxia tolerance in fish.
Collapse
Affiliation(s)
- Qiang Ma
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China
| | - Houguo Xu
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, China
| | - Yuliang Wei
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, China
| | - Mengqing Liang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, China.
| |
Collapse
|
37
|
Rachman MP, Bamidele O, Dessie T, Smith J, Hanotte O, Gheyas AA. Genomic analysis of Nigerian indigenous chickens reveals their genetic diversity and adaptation to heat-stress. Sci Rep 2024; 14:2209. [PMID: 38278850 PMCID: PMC10817956 DOI: 10.1038/s41598-024-52569-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/20/2024] [Indexed: 01/28/2024] Open
Abstract
Indigenous poultry breeds from Africa can survive in harsh tropical environments (such as long arid seasons, excessive rain and humidity, and extreme heat) and are resilient to disease challenges, but they are not productive compared to their commercial counterparts. Their adaptive characteristics are in response to natural selection or to artificial selection for production traits that have left selection signatures in the genome. Identifying these signatures of positive selection can provide insight into the genetic bases of tropical adaptations observed in indigenous poultry and thereby help to develop robust and high-performing breeds for extreme tropical climates. Here, we present the first large-scale whole-genome sequencing analysis of Nigerian indigenous chickens from different agro-climatic conditions, investigating their genetic diversity and adaptation to tropical hot climates (extreme arid and extreme humid conditions). The study shows a large extant genetic diversity but low level of population differentiation. Using different selection signature analyses, several candidate genes for adaptation were detected, especially in relation to thermotolerance and immune response (e.g., cytochrome P450 2B4-like, TSHR, HSF1, CDC37, SFTPB, HIF3A, SLC44A2, and ILF3 genes). These results have important implications for conserving valuable genetic resources and breeding improvement of chickens for thermotolerance.
Collapse
Affiliation(s)
- Mifta P Rachman
- School of Biosciences, University of Nottingham, Nottingham, LE12 5RD, UK.
| | - Oladeji Bamidele
- African Chicken Genetic Gains (ACGG), Department of Animal Sciences, Obafemi Awolowo University, Ile Ife, 220282, Nigeria
| | - Tadelle Dessie
- LiveGene-CTLGH, International Livestock Research Institute (ILRI), P.O. Box 5689, Addis Ababa, Ethiopia
| | - Jacqueline Smith
- Centre for Tropical Livestock Genetics and Health (CTLGH), Roslin Institute, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Olivier Hanotte
- LiveGene-CTLGH, International Livestock Research Institute (ILRI), P.O. Box 5689, Addis Ababa, Ethiopia.
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2RD, UK.
| | - Almas A Gheyas
- Centre for Tropical Livestock Genetics and Health (CTLGH), Roslin Institute, University of Edinburgh, Edinburgh, EH25 9RG, UK.
| |
Collapse
|
38
|
Malovic E, Ealy A, Hsu PJ, Sarkar S, Miller C, Rokad D, Goeser C, Hartman AK, Zhu A, Palanisamy B, Zenitsky G, Jin H, Anantharam V, Kanthasamy A, He C, Kanthasamy AG. Epitranscriptomic Reader YTHDF2 Regulates SEK1( MAP2K4 )-JNK-cJUN Inflammatory Signaling in Astrocytes during Neurotoxic Stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.26.577106. [PMID: 38328119 PMCID: PMC10849634 DOI: 10.1101/2024.01.26.577106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
As the most abundant glial cells in the CNS, astrocytes dynamically respond to neurotoxic stress, however, the key molecular regulators controlling the inflammatory status of these sentinels during neurotoxic stress have remained elusive. Herein, we demonstrate that the m6A epitranscriptomic mRNA modification tightly regulates the pro-inflammatory functions of astrocytes. Specifically, the astrocytic neurotoxic stresser, manganese (Mn), downregulated the m6A reader YTHDF2 in human and mouse astrocyte cultures and in the mouse brain. Functionally, YTHDF2 knockdown augmented, while its overexpression dampened, neurotoxic stress induced proinflammatory response, suggesting YTHDF2 serves as a key upstream regulator of inflammatory responses in astrocytes. Mechnistically, YTHDF2 RIP-sequencing identified MAP2K4 ( MKK4; SEK1) mRNA as a YTHDF2 target influencing inflammatory signaling. Our target validation revealed Mn-exposed astrocytes mediates proinflammatory response by activating the phosphorylation of SEK1, JNK, and cJUN signaling. Collectively, YTHDF2 serves a key upstream 'molecular switch' controlling SEK1( MAP2K4 )-JNK-cJUN proinflammatory signaling in astrocytes.
Collapse
|
39
|
Qiu Y, Zeng Y, Zhang C, Lv X, Ling Y, Si Y, Guo T, Ni Y, Zhang J, Xu C, Wang Z, Hu J. A ROS-responsive loaded desferoxamine (DFO) hydrogel system for traumatic brain injury therapy. Biomed Mater 2024; 19:025016. [PMID: 38215474 DOI: 10.1088/1748-605x/ad1dfd] [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: 07/13/2023] [Accepted: 01/12/2024] [Indexed: 01/14/2024]
Abstract
Traumatic brain injury (TBI) produces excess iron, and increased iron accumulation in the brain leads to lipid peroxidation and reactive oxygen species (ROSs), which can exacerbate secondary damage and lead to disability and death. Therefore, inhibition of iron overload and oxidative stress has a significant role in the treatment of TBI. Functionalized hydrogels with iron overload inhibiting ability and of oxidative stress inhibiting ability will greatly contribute to the repair of TBI. Herein, an injectable, post-traumatic microenvironment-responsive, ROS-responsive hydrogel encapsulated with deferrioxamine mesylate (DFO) was developed. The hydrogel is rapidly formed via dynamic covalent bonding between phenylboronic acid grafted hyaluronic acid (HA-PBA) and polyvinyl alcohol (PVA), and phenylboronate bonds are used to respond to and reduce ROS levels in damaged brain tissue to promote neuronal recovery. The release of DFO from HA-PBA/PVA hydrogels in response to ROS further promotes neuronal regeneration and recovery by relieving iron overload and thus eradicating ROS. In the Feeney model of Sprague Dawley rats, HA-PBA/PVA/DFO hydrogel treatment significantly improved the behavior of TBI rats and reduced the area of brain contusion in rats. In addition, HA-PBA/PVA/DFO hydrogel significantly reduced iron overload to reduce ROS and could effectively promote post-traumatic neuronal recovery. Its effects were also explored, and notably, HA-PBA/PVA/DFO hydrogel can reduce iron overload as well as ROS, thus protecting neurons from death. Thus, this injectable, biocompatible and ROS-responsive drug-loaded hydrogel has great potential for the treatment of TBI. This work suggests a novel method for the treatment of secondary brain injury by inhibiting iron overload and the oxidative stress response after TBI.
Collapse
Affiliation(s)
- Yun Qiu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Yu Zeng
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Chun Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Xiaorui Lv
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Yating Ling
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Yu Si
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Tao Guo
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Yinying Ni
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Jingwen Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Changgen Xu
- Zhenjiang Blood Center, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Ziyu Wang
- Health Clinical Laboratories, Health BioMed Co., Ltd, Ningbo, Zhejiang 315042, People's Republic of China
| | - Jiabo Hu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
- Zhenjiang Blood Center, Zhenjiang, Jiangsu 212013, People's Republic of China
| |
Collapse
|
40
|
Sri Hari A, Metcalf CS. The "Curious Case of Postictal Oxygen Dissipation": Could ROS Be the Culprit? Epilepsy Curr 2024; 24:59-61. [PMID: 38327542 PMCID: PMC10846519 DOI: 10.1177/15357597231216921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024] Open
Abstract
Postictal Hypoxia Involves Reactive Oxygen Species and Is Ameliorated by Chronic Mitochondrial Uncoupling Villa BR, George AG, Shutt TE, Sullivan PG, Rho JM, Teskey GC. Neuropharmacology . 2023;238:109653. doi:10.1016/j.neuropharm.2023.109653 Prolonged severe hypoxia follows brief seizures and represents a mechanism underlying several negative postictal manifestations without interventions. Approximately 50% of the postictal hypoxia phenomenon can be accounted for by arteriole vasoconstriction. What accounts for the rest of the drop in unbound oxygen is unclear. Here, we determined the effect of pharmacological modulation of mitochondrial function on tissue oxygenation in the hippocampus of rats after repeatedly evoked seizures. Rats were treated with mitochondrial uncoupler 2,4 dinitrophenol (DNP) or antioxidants. Oxygen profiles were recorded using a chronically implanted oxygen-sensing probe, before, during, and after seizure induction. Mitochondrial function and redox tone were measured using in vitro mitochondrial assays and immunohistochemistry. Postictal cognitive impairment was assessed using the novel object recognition task. Mild mitochondrial uncoupling by DNP raised hippocampal oxygen tension and ameliorated postictal hypoxia. Chronic DNP also lowered mitochondrial oxygen-derived reactive species and oxidative stress in the hippocampus during postictal hypoxia. Uncoupling the mitochondria exerts therapeutic benefits on postictal cognitive dysfunction. Finally, antioxidants do not affect postictal hypoxia, but protect the brain from associated cognitive deficits. We provided evidence for a metabolic component of the prolonged oxygen deprivation that follow seizures and its pathological sequelae. Furthermore, we identified a molecular underpinning of this metabolic component, which involves excessive oxygen conversion into reactive species. Mild mitochondrial uncoupling may be a potential therapeutic strategy to treat the postictal state where seizure control is absent or poor.
Collapse
|
41
|
Barakat RM, Turcani M, Al-Khaledi G, Kilarkaje N, Al-Sarraf H, Sayed Z, Redzic Z. Low oxygen in inspired air causes severe cerebrocortical hypoxia and cell death in the cerebral cortex of awake rats. Neurosci Lett 2024; 818:137515. [PMID: 37865187 DOI: 10.1016/j.neulet.2023.137515] [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: 03/12/2023] [Revised: 09/12/2023] [Accepted: 10/09/2023] [Indexed: 10/23/2023]
Abstract
Type 1 respiratory failure (T1RF) is associated with secondary acute brain injury (sABI). The underlying mechanisms of sABI could include injury to brain cells mediated either by hypoxia or by lung injury-triggered inflammation. To elucidate to what extent T1RF causes hypoxia and a consequent hypoxic injury in the brain in the absence of lung injury, we exposed healthy, conscious Sprague-Dawley rats to 48 h long low partial pressure of O2 in inspired air (PiO2) (7.5-8 % O2 in N2, CO2 < 0.5 %, normal barometric pressure) and measured the partial pressure of oxygen in the premotor cortex (PtO2), cerebral blood flow (CBF), lactate concentrations, and cell death. Low PiO2 significantly affected PtO2, which was 52.3 (SD 2.1) mmHg when PiO2 was normal but declined to 6.4 (SD 3.8) mmHg when PiO2 was low for 1 h. This was accompanied by increased lactate concentrations in plasma, CSF, and premotor cortex. Low PiO2 elevated the number of dead cells in the cerebral cortex from 5.6 (SD 4.8) % (when PiO2 was normal) to 20.5 (SD 4.1) % and 32.37 (SD 6.5) % after 24 h and 48 h exposure to low PiO2, respectively. The Mann-Kendall test could not detect any monotonic increase or decrease in pial blood flow during the 48 h exposure to low PiO2. In summary, our findings suggest that exposure to low PiO2 caused a severe hypoxia in the cerebral cortex, which triggers a massive cell death. Since these conditions mimic T1RF, hypoxic injury could be an important underlying cause of T1RF-induced sABI.
Collapse
Affiliation(s)
- Rawan M Barakat
- Department of Physiology, College of Medicine, Kuwait University, Kuwait
| | - Marian Turcani
- Department of Physiology, College of Medicine, Kuwait University, Kuwait
| | - Ghanim Al-Khaledi
- Department of Pharmacology, College of Medicine, Kuwait University, Kuwait
| | | | - Hameed Al-Sarraf
- Department of Physiology, College of Medicine, Kuwait University, Kuwait
| | - Zeinab Sayed
- Department of Physiology, College of Medicine, Kuwait University, Kuwait
| | - Zoran Redzic
- Department of Physiology, College of Medicine, Kuwait University, Kuwait.
| |
Collapse
|
42
|
Damgaard V, Mariegaard J, Lindhardsen JM, Ehrenreich H, Miskowiak KW. Neuroprotective Effects of Moderate Hypoxia: A Systematic Review. Brain Sci 2023; 13:1648. [PMID: 38137096 PMCID: PMC10741927 DOI: 10.3390/brainsci13121648] [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: 10/31/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Emerging evidence highlights moderate hypoxia as a candidate treatment for brain disorders. This systematic review examines findings and the methodological quality of studies investigating hypoxia (10-16% O2) for ≥14 days in humans, as well as the neurobiological mechanisms triggered by hypoxia in animals, and suggests optimal treatment protocols to guide future studies. We followed the preferred reporting items for systematic reviews and meta-analysis (PRISMA) 2020. Searches were performed on PubMed/MEDLINE, PsycInfo, EMBASE, and the Cochrane Library, in May-September 2023. Two authors independently reviewed the human studies with the following tools: (1) revised Cochrane collaboration's risk of bias for randomized trials 2.0; (2) the risk of bias in nonrandomized studies of interventions. We identified 58 eligible studies (k = 8 human studies with N = 274 individuals; k = 48 animal studies) reporting the effects of hypoxia on cognition, motor function, neuroimaging, neuronal/synaptic morphology, inflammation, oxidative stress, erythropoietin, neurotrophins, and Alzheimer's disease markers. A total of 75% of human studies indicated cognitive and/or neurological benefits, although all studies were evaluated ashigh risk of bias due to a lack of randomization and assessor blinding. Low-dose intermittent or continuous hypoxia repeated for 30-240 min sessions, preferably in combination with motor-cognitive training, produced beneficial effects, and high-dose hypoxia with longer (≥6 h) durations and chronic exposure produced more adverse effects. Larger and methodologically stronger translational studies are warranted.
Collapse
Affiliation(s)
- Viktoria Damgaard
- Neurocognition and Emotion in Affective Disorders (NEAD) Centre, Copenhagen Affective Disorder Research Centre, Psychiatric Centre Copenhagen, Frederiksberg Hospital, Hovedvejen 17, DK-2000 Frederiksberg, Denmark; (V.D.); (J.M.)
- Department of Psychology, University of Copenhagen, Øster Farimagsgade 2A, DK-1353 Copenhagen, Denmark
| | - Johanna Mariegaard
- Neurocognition and Emotion in Affective Disorders (NEAD) Centre, Copenhagen Affective Disorder Research Centre, Psychiatric Centre Copenhagen, Frederiksberg Hospital, Hovedvejen 17, DK-2000 Frederiksberg, Denmark; (V.D.); (J.M.)
- Department of Psychology, University of Copenhagen, Øster Farimagsgade 2A, DK-1353 Copenhagen, Denmark
| | - Julie Marie Lindhardsen
- Neurocognition and Emotion in Affective Disorders (NEAD) Centre, Copenhagen Affective Disorder Research Centre, Psychiatric Centre Copenhagen, Frederiksberg Hospital, Hovedvejen 17, DK-2000 Frederiksberg, Denmark; (V.D.); (J.M.)
- Department of Psychology, University of Copenhagen, Øster Farimagsgade 2A, DK-1353 Copenhagen, Denmark
| | - Hannelore Ehrenreich
- University of Göttingen, 37075 Göttingen, Germany;
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany
| | - Kamilla Woznica Miskowiak
- Neurocognition and Emotion in Affective Disorders (NEAD) Centre, Copenhagen Affective Disorder Research Centre, Psychiatric Centre Copenhagen, Frederiksberg Hospital, Hovedvejen 17, DK-2000 Frederiksberg, Denmark; (V.D.); (J.M.)
- Department of Psychology, University of Copenhagen, Øster Farimagsgade 2A, DK-1353 Copenhagen, Denmark
| |
Collapse
|
43
|
Liao Y, Chen Z, Yang Y, Shen D, Chai S, Ma Y, Ge R, Wang X, Wang S, Liu S. Antibiotic intervention exacerbated oxidative stress and inflammatory responses in SD rats under hypobaric hypoxia exposure. Free Radic Biol Med 2023; 209:70-83. [PMID: 37806597 DOI: 10.1016/j.freeradbiomed.2023.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/28/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
The gut microbiota plays a crucial role in maintaining host nutrition, metabolism, and immune homeostasis, particularly in extreme environmental conditions. However, the regulatory mechanisms of the gut microbiota in animal organisms hypobaric hypoxia exposure require further study. We conducted a research by comparing SD rats treated with an antibiotic (ABX) cocktail and untreated SD rats that were housed in a low-pressure oxygen chamber (simulating low pressure and hypoxic environment at 6000 m altitude) for 30 days. After the experiment, blood, feces, and lung tissues from SD rats were collected for analysis of blood, 16S rRNA amplicon sequencing, and non-targeted metabolomics. The results demonstrated that the antibiotic cocktail-treated SD rats exhibited elevated counts of neutrophil (Neu) and monocyte (Mon) cells, an enrichment of sulfate-reducing bacteria (SBC), reduced levels of glutathione, and accumulated phospholipid compounds. Notably, the accumulation of phospholipid compounds, particularly lysophosphatidic acid (LPA), lipopolysaccharide (LPS), and lysophosphatidylcholine (LPC), along with the aforementioned changes, contributed to heightened oxidative stress and inflammation in the organism. In addition, we explored the resistance mechanisms of SD rats in low-oxygen and low-pressure environments and found that increasing the quantity of the Prevotellaceae and related beneficial bacteria (especially Lactobacillus) could reduce oxidative stress and inflammation. These findings offer valuable insights into enhancing the adaptability of low-altitude animals under hypobaric hypoxia exposure.
Collapse
Affiliation(s)
- Yang Liao
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China
| | - Zheng Chen
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China
| | - Yingkui Yang
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China
| | - Di Shen
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China
| | - Shatuo Chai
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China
| | - Yan Ma
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, 810001, China
| | - Rili Ge
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, 810001, China
| | - Xun Wang
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China
| | - Shuxiang Wang
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China.
| | - Shujie Liu
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China.
| |
Collapse
|
44
|
Villa BR, George AG, Shutt TE, Sullivan PG, Rho JM, Teskey GC. Postictal hypoxia involves reactive oxygen species and is ameliorated by chronic mitochondrial uncoupling. Neuropharmacology 2023; 238:109653. [PMID: 37422182 DOI: 10.1016/j.neuropharm.2023.109653] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
Prolonged severe hypoxia follows brief seizures and represents a mechanism underlying several negative postictal manifestations without interventions. Approximately 50% of the postictal hypoxia phenomenon can be accounted for by arteriole vasoconstriction. What accounts for the rest of the drop in unbound oxygen is unclear. Here, we determined the effect of pharmacological modulation of mitochondrial function on tissue oxygenation in the hippocampus of rats after repeatedly evoked seizures. Rats were treated with mitochondrial uncoupler 2,4 dinitrophenol (DNP) or antioxidants. Oxygen profiles were recorded using a chronically implanted oxygen-sensing probe, before, during, and after seizure induction. Mitochondrial function and redox tone were measured using in vitro mitochondrial assays and immunohistochemistry. Postictal cognitive impairment was assessed using the novel object recognition task. Mild mitochondrial uncoupling by DNP raised hippocampal oxygen tension and ameliorated postictal hypoxia. Chronic DNP also lowered mitochondrial oxygen-derived reactive species and oxidative stress in the hippocampus during postictal hypoxia. Uncoupling the mitochondria exerts therapeutic benefits on postictal cognitive dysfunction. Finally, antioxidants do not affect postictal hypoxia, but protect the brain from associated cognitive deficits. We provided evidence for a metabolic component of the prolonged oxygen deprivation that follow seizures and its pathological sequelae. Furthermore, we identified a molecular underpinning of this metabolic component, which involves excessive oxygen conversion into reactive species. Mild mitochondrial uncoupling may be a potential therapeutic strategy to treat the postictal state where seizure control is absent or poor.
Collapse
Affiliation(s)
- Bianca R Villa
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, T2N 4N1, Canada.
| | - Antis G George
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, T2N 4N1, Canada.
| | - Timothy E Shutt
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada; Departments of Medical Genetics and Biochemistry & Molecular Biology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada; Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada.
| | - Patrick G Sullivan
- Department of Anatomy and Neurobiology, and Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, 40536, USA.
| | - Jong M Rho
- Department of Neurosciences, Pediatrics and Pharmacology, University of California San Diego, Rady Children's Hospital, San Diego, CA, 92037, USA.
| | - G Campbell Teskey
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, T2N 4N1, Canada; Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
45
|
Tang L, Wang Y, Xiang J, Yang D, Zhang Y, Xiang Q, Li J. lncRNA and circRNA expression profiles in the hippocampus of Aβ 25‑35‑induced AD mice treated with Tripterygium glycoside. Exp Ther Med 2023; 26:426. [PMID: 37602300 PMCID: PMC10433443 DOI: 10.3892/etm.2023.12125] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 03/15/2023] [Indexed: 08/22/2023] Open
Abstract
Tripterygium glycosides (TG) have been reported to ameliorate Alzheimer's disease (AD), although the mechanism involved remains to be determined. In the present study, the lncRNA and circRNA expression profiles of an AD mouse model treated with TG were assessed using microarrays. lncRNAs, mRNAs, and circRNAs in the hippocampi of 3 AD+normal saline (NS) mice and 3 AD+TG mice were detected using microarrays. The most differentially expressed lncRNAs, mRNAs, and circRNAs were screened between the AD+NS and AD+TG groups. The differentially expressed lncRNAs and circRNAs were analyzed using GO enrichment and KEGG analyses. Co-expression analysis of lncRNAs, circRNAs, and mRNAs was performed by calculating the correlation coefficients. Protein-protein interaction (PPI) network analysis was performed on mRNAs using STRING. The lncRNA-target-transcription factor (TF) network was analyzed using the Network software. In total, 661 lncRNAs, 64 circRNAs, and 503 mRNAs were found to be differentially expressed in AD mice treated with TG. Pou4f1, Egr2, Mag, and Nr4a1 were the hub genes in the PPI network. The KEGG results showed that the mRNAs that were co-expressed with lncRNAs were enriched in the TNF, PI3K-Akt, and Wnt signaling pathways. LncRNA-target-TF network analysis indicated that TFs, including Cebpa, Zic2, and Rxra, were the most likely to regulate the detected lncRNAs. The circRNA-miRNA interaction network indicated that 275 miRNAs may bind to the 64 circRNAs. In conclusion, these findings provide a novel perspective on AD pathogenesis, and the detected lncRNAs, mRNAs, and circRNAs may serve as novel therapeutic targets for the management of AD.
Collapse
Affiliation(s)
- Liang Tang
- Department of Basic Biology, Changsha Medical College, Changsha, Hunan 410219, P.R. China
- Department of Basic Biology, Wuzhou Medical College, Wuzhou, Guangxi Zhuang 543000, P.R. China
- Center for Neuroscience and Behavior, Changsha Medical College, Changsha, Hunan 410219, P.R. China
- The Hunan Provincial University Key Laboratory of The Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical College, Changsha, Hunan 410219, P.R. China
| | - Yan Wang
- Department of Basic Biology, Changsha Medical College, Changsha, Hunan 410219, P.R. China
- Department of Basic Biology, Wuzhou Medical College, Wuzhou, Guangxi Zhuang 543000, P.R. China
- Center for Neuroscience and Behavior, Changsha Medical College, Changsha, Hunan 410219, P.R. China
| | - Ju Xiang
- Department of Basic Biology, Changsha Medical College, Changsha, Hunan 410219, P.R. China
- Center for Neuroscience and Behavior, Changsha Medical College, Changsha, Hunan 410219, P.R. China
- The Hunan Provincial University Key Laboratory of The Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical College, Changsha, Hunan 410219, P.R. China
| | - Dawei Yang
- Department of Basic Biology, Changsha Medical College, Changsha, Hunan 410219, P.R. China
- Center for Neuroscience and Behavior, Changsha Medical College, Changsha, Hunan 410219, P.R. China
- The Hunan Provincial University Key Laboratory of The Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical College, Changsha, Hunan 410219, P.R. China
| | - Yan Zhang
- Department of Basic Biology, Changsha Medical College, Changsha, Hunan 410219, P.R. China
- Center for Neuroscience and Behavior, Changsha Medical College, Changsha, Hunan 410219, P.R. China
- The Hunan Provincial University Key Laboratory of The Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical College, Changsha, Hunan 410219, P.R. China
- School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, P.R. China
| | - Qin Xiang
- Department of Basic Biology, Changsha Medical College, Changsha, Hunan 410219, P.R. China
- Center for Neuroscience and Behavior, Changsha Medical College, Changsha, Hunan 410219, P.R. China
- The Hunan Provincial University Key Laboratory of The Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical College, Changsha, Hunan 410219, P.R. China
| | - Jianming Li
- Department of Basic Biology, Changsha Medical College, Changsha, Hunan 410219, P.R. China
- Center for Neuroscience and Behavior, Changsha Medical College, Changsha, Hunan 410219, P.R. China
- The Hunan Provincial University Key Laboratory of The Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical College, Changsha, Hunan 410219, P.R. China
| |
Collapse
|
46
|
Dakterzada F, Benítez ID, Targa A, Carnes A, Pujol M, Jové M, Mínguez O, Vaca R, Sánchez-de-la-Torre M, Barbé F, Pamplona R, Piñol-Ripoll G. Cerebrospinal fluid lipidomic fingerprint of obstructive sleep apnoea in Alzheimer's disease. Alzheimers Res Ther 2023; 15:134. [PMID: 37550750 PMCID: PMC10408111 DOI: 10.1186/s13195-023-01278-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 07/21/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND Obstructive sleep apnoea (OSA) has a high prevalence in patients with Alzheimer's disease (AD). Both conditions have been shown to be associated with lipid dysregulation. However, the relationship between OSA severity and alterations in lipid metabolism in the brains of patients with AD has yet to be fully elucidated. In this context, we examined the cerebrospinal fluid (CSF) lipidome of patients with suspected OSA to identify potential diagnostic biomarkers and to provide insights into the pathophysiological mechanisms underlying the effect of OSA on AD. METHODS The study included 91 consecutive AD patients who underwent overnight polysomnography (PSG) to diagnose severe OSA (apnoea-hypopnea index ≥ 30/h). The next morning, CSF samples were collected and analysed by liquid chromatography coupled to mass spectrometry in an LC-ESI-QTOF-MS/MS platform. RESULTS The CSF levels of 11 lipid species were significantly different between AD patients with (N = 38) and without (N = 58) severe OSA. Five lipids (including oxidized triglyceride OxTG(57:2) and four unknown lipids) were significantly correlated with specific PSG measures of OSA severity related to sleep fragmentation and hypoxemia. Our analyses revealed a 4-lipid signature (including oxidized ceramide OxCer(40:6) and three unknown lipids) that provided an accuracy of 0.80 (95% CI: 0.71-0.89) in the detection of severe OSA. These lipids increased the discriminative power of the STOP-Bang questionnaire in terms of the area under the curve (AUC) from 0.61 (0.50-0.74) to 0.85 (0.71-0.93). CONCLUSIONS Our results reveal a CSF lipidomic fingerprint that allows the identification of AD patients with severe OSA. Our findings suggest that an increase in central nervous system lipoxidation may be the principal mechanism underlying the association between OSA and AD.
Collapse
Affiliation(s)
- Farida Dakterzada
- Unitat Trastorns Cognitius, Cognition and Behaviour Study Group, Santa Maria University Hospital, IRBLleida, Rovira Roure No. 44, Lleida, 25198, Spain
| | - Iván D Benítez
- Group of Translational Research in Respiratory Medicine, Hospital Universitari Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
- Center for Biomedical Research in Respiratory Diseases Network (CIBERES), Madrid, Spain
| | - Adriano Targa
- Group of Translational Research in Respiratory Medicine, Hospital Universitari Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
- Center for Biomedical Research in Respiratory Diseases Network (CIBERES), Madrid, Spain
| | - Anna Carnes
- Unitat Trastorns Cognitius, Cognition and Behaviour Study Group, Santa Maria University Hospital, IRBLleida, Rovira Roure No. 44, Lleida, 25198, Spain
| | - Montse Pujol
- Group of Translational Research in Respiratory Medicine, Hospital Universitari Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida (UdL-IRBLleida), Lleida, Spain
| | - Olga Mínguez
- Group of Translational Research in Respiratory Medicine, Hospital Universitari Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
| | - Rafi Vaca
- Group of Translational Research in Respiratory Medicine, Hospital Universitari Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
| | - Manuel Sánchez-de-la-Torre
- Department of Nursing and Physiotherapy, Group of Precision Medicine in Chronic Diseases, University Hospital Arnau de Vilanova and Santa María, IRBLleida, Faculty of Nursing and Physiotherapy, University of Lleida, Lleida, Spain
| | - Ferran Barbé
- Group of Translational Research in Respiratory Medicine, Hospital Universitari Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
- Center for Biomedical Research in Respiratory Diseases Network (CIBERES), Madrid, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida (UdL-IRBLleida), Lleida, Spain
| | - Gerard Piñol-Ripoll
- Unitat Trastorns Cognitius, Cognition and Behaviour Study Group, Santa Maria University Hospital, IRBLleida, Rovira Roure No. 44, Lleida, 25198, Spain.
| |
Collapse
|
47
|
Ma JL, Xu DP, Tao YF, Zheng T, Xu P, Qiang J. Integrated transcriptome and miRNA sequencing analyses reveal that hypoxia stress induces immune and metabolic disorders in gill of genetically improved farmed tilapia (GIFT, Oreochromis niloticus). FISH & SHELLFISH IMMUNOLOGY 2023; 139:108909. [PMID: 37353064 DOI: 10.1016/j.fsi.2023.108909] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/12/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023]
Abstract
The survival and growth of fish are significantly impacted by a hypoxic environment (low dissolved oxygen). In this study, we compared tissue structure, physiological changes, and mRNA/miRNA transcriptome, in gills of genetically improved farmed tilapia (GIFT, Oreochromis niloticus) between the hypoxic group (DO: 0.55 mg/L, HG) and the control group (DO: 5 mg/L, CG). The results showed that the gill filaments in the hypoxic group showed curling, engorgement, and apoptotic cells increased, and that exposure for 96 h resulted in a reduction in the antioxidant capacity. We constructed and sequenced miRNA and mRNA libraries from gill tissues of GIFT at 96 h of hypoxia stress. Between the HG and CG, a total of 14 differentially expressed (DE) miRNAs and 1557 DE genes were obtained. GO and KEGG enrichment showed that DE genes were mainly enriched in immune and metabolic pathways such as natural killer cell mediated cytotoxicity, steroid biosynthesis, primary immunodeficiency, and synthesis and degradation of ketone bodies. Based on the results of mRNA sequencing and screening for miRNA-mRNA pairs, we selected and verified six DE miRNAs and their probable target genes. The sequencing results were consistent with the qRT-PCR validation results. The result showed that under hypoxia stress, the innate immune response was up-regulated, and the adaptive immune response was down-regulated in the gill of GIFT. The synthesis of cholesterol in gill cells is reduced, which is conducive to the absorption of solvent oxygen. These findings offer fresh information about the processes of fish adaptation to hypoxic stress.
Collapse
Affiliation(s)
- Jun-Lei Ma
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Dong-Po Xu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China; Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China.
| | - Yi-Fan Tao
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Tao Zheng
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Pao Xu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China; Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Jun Qiang
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China; Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China.
| |
Collapse
|
48
|
Wang Y, Zhang W, Xu G, Shi C, Wang X, Qu J, Wang H, Liu C. The role of TRPV4 in the regulation of retinal ganglion cells apoptosis in rat and mouse. Heliyon 2023; 9:e17583. [PMID: 37456002 PMCID: PMC10338314 DOI: 10.1016/j.heliyon.2023.e17583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
Retinal ganglion cell (RGC) damages are common in glaucoma, causing atrophy of the optic papilla, visual field damage, and visual loss. Transient receptor potential vanilloid 4 (TRPV4) is significantly expressed in the eyeball and is sensitive to mechanical and osmotic pressure. However, the specific role and mechanism of TRPV4 in glaucoma and RGC progression remain unclear. TRPV4 expression was detected in RGCs under different pressure culture conditions. We also explored the pressure effect on TRPV4 expression and the role and mechanism behind the functional regulation of RGCs. Immunofluorescence staining, western blotting, and TUNEL were utilized in this study. Our results established that TRPV4 was expressed in RGCs. TRPV4 expression was decreased at 40 mmHg and 60 mmHg, and the expression of BAX at 40 mmHg, 60 mmHg. Additionally, the expression of caspase 9 protein increased at 40 mmHg with the pressure increase compared with the conventional culture group. TUNEL staining revealed that the apoptosis rate of RGCs was elevated at 40 mmHg and 60 mmHg, compared with the traditional culture group. Therefore, the expression of BAX and caspase 9 increased, along with the apoptosis rate of RGCs compared with the control group. However, after TRPV4 antagonist treatment, the expression of BAX and caspase 9 decreased, and the apoptosis rate of RGCs decreased. Thus, TRPV4 may affect the mitochondrial apoptosis pathway, such as BAX and caspase 9, leading to the apoptosis of RGCs. The antagonists of TRPV4 could provide a new idea for clinically treating acute glaucoma.
Collapse
Affiliation(s)
- Yi Wang
- Department of Ophthalmology, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, 271000, China
| | - Wei Zhang
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Guozheng Xu
- Department of Physiology and Neurobiology, Functional Laboratory, Shandong First Medical University, Jinan, Shandong, 250117, China
| | - Changwei Shi
- Department of Physiology and Neurobiology, Functional Laboratory, Shandong First Medical University, Jinan, Shandong, 250117, China
| | - Xiang Wang
- School of Clinical Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271016, China
| | - Jianfeng Qu
- Medical Engineering and Technology Research Center, Shandong First Medical University, Taian, Shandong, 271000, China
| | - Hongmei Wang
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Chunhua Liu
- Department of Physiology and Neurobiology, Functional Laboratory, Shandong First Medical University, Jinan, Shandong, 250117, China
| |
Collapse
|
49
|
Wen Q, Hong X, He K, Liu B, Li M. Can acupuncture reverse oxidative stress and neuroinflammatory damage in animal models of vascular dementia?: A preclinical systematic review and meta-analysis. Medicine (Baltimore) 2023; 102:e33989. [PMID: 37335660 PMCID: PMC10256398 DOI: 10.1097/md.0000000000033989] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/23/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Vascular dementia is a cognitive dysfunction syndrome caused by cerebral vascular factors such as ischemic stroke and hemorrhagic stroke. The effect of acupuncture on vascular dementia models is ambiguous, and there is controversy about whether acupuncture has a placebo effect. Oxidative stress and inflammation are the most essential mechanisms in preclinical studies of vascular dementia. However, there is no meta-analysis on the mechanism of vascular dementia in animal models. It is necessary to explore the efficacy of acupuncture through Meta-analysis of preclinical studies. METHODS Three major databases, PubMed, Embase and Web of Science (including medline), were searched in English until December 2022.The quality of the including literature was assessed using SYRCLE's risk of bias tool. Review Manager 5.3 was used to statistically summarize the included studies and the statistical effect values were expressed by SMD. The outcomes included: behavioral tests (escape latency, number of crossings), pathological sections (Nissl and TUNEL staining), oxidative stress markers (ROS, MDA, SOD, GSH-PX) and neuroinflammatory factors (TNF-α, IL-1β, IL-6). RESULTS A total of 31 articles were included in this meta-analysis. The results showed that the escape latency, the contents of ROS, MDA, IL-1β, and IL-6 were decreased, and the contents of SOD and Nissl-positive neurons were increased in the acupuncture group as compared with the non-group (P < .05). Compared with the impaired group, the acupuncture group also had the above advantages (P < .05). In addition, the acupuncture group also increased the number of crossings and GSH-PX content, and decreased the expression of TUNEL-positive neurons and TNF-α (P < .05). CONCLUSIONS From behavioral tests to slices and pathological markers in animal models of vascular dementia, it can be proved that acupuncture is effective in targeting oxidative stress and neuroinflammatory damage, and acupuncture is not a placebo effect. Nevertheless, attention needs to be paid to the gap between animal experiments and clinical applications.
Collapse
Affiliation(s)
- Qin Wen
- Clinical Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
- Yuangang Street Community Health Service Center, Guangzhou, China
| | - Xueqin Hong
- Clinical Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kunze He
- Clinical Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Buping Liu
- Clinical Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min Li
- Clinical Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
50
|
Li XL, Liu YL, Liu JY, Zhu YY, Zhu XX, Zhang WW, Li J, Zhao Y, Zhao LL, Zhang C, Wang H, Xu DX, Gao L. 1-Nitropyrene disrupts testicular steroidogenesis via oxidative stress-evoked PERK-eIF2α pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 259:115027. [PMID: 37207578 DOI: 10.1016/j.ecoenv.2023.115027] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/21/2023]
Abstract
Our previous study showed 1-Nitropyrene (1-NP) exposure disrupted testicular testosterone synthesis in mouse, but the exact mechanism needs further investigation. The present research found 4-phenylbutyric acid (4-PBA), an endoplasmic reticulum (ER) stress inhibitor, recovered 1-NP-induced ER stress and testosterone synthases reduction in TM3 cells. GSK2606414, a protein kinase-like ER kinase (PERK) kinase inhibitor, attenuated 1-NP-induced PERK-eukaryotic translation initiation factor 2α (eIF2α) signaling activation and downregulation of steroidogenic proteins in TM3 cells. Both 4-PBA and GSK2606414 attenuated 1-NP-induced steroidogenesis disruption in TM3 cells. Further studies used N-Acetyl-L-cysteine (NAC) as a classical antioxidant to explore whether oxidative stress-activated ER stress mediated 1-NP-induced testosterone synthases reduction and steroidogenesis disruption in TM3 cells and mouse testes. The results showed NAC pretreatment mitigated oxidative stress, and subsequently attenuated ER stress, particularly PERK-eIF2α signaling activation, and downregulation of testosterone synthases in 1-NP-treated TM3 cells. More importantly, NAC extenuated 1-NP-induced testosterone synthesis in vitro and in vivo. The current work indicated that oxidative stress-caused ER stress, particularly PERK-eIF2α pathway activation, mediates 1-NP-downregulated steroidogenic proteins and steroidogenesis disruption in TM3 cells and mouse testes. Significantly, the current study provides a theoretical basis and demonstrates the experimental evidence for the potential application of antioxidant, such as NAC, in public health prevention, particularly in 1-NP-induced endocrine disorder.
Collapse
Affiliation(s)
- Xiu-Liang Li
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Yu-Lin Liu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Jia-Yu Liu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Yan-Yan Zhu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Xin-Xin Zhu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Wei-Wei Zhang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Jian Li
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Ye Zhao
- Department of Nuclear Medicine, Anhui Medical University, Hefei 230032, China
| | - Ling-Li Zhao
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Cheng Zhang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Hua Wang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - De-Xiang Xu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China.
| | - Lan Gao
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|