1
|
Tregub PP, Komleva YK, Kulikov VP, Chekulaev PA, Tregub OF, Maltseva LD, Manasova ZS, Popova IA, Andriutsa NS, Samburova NV, Salmina AB, Litvitskiy PF. Relationship between Hypoxia and Hypercapnia Tolerance and Life Expectancy. Int J Mol Sci 2024; 25:6512. [PMID: 38928217 PMCID: PMC11204369 DOI: 10.3390/ijms25126512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/05/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
The review discusses the potential relationship between hypoxia resistance and longevity, the influence of carbon dioxide on the mechanisms of aging of the mammalian organism, and intermittent hypercapnic-hypoxic effects on the signaling pathways of aging mechanisms. In the article, we focused on the potential mechanisms of the gero-protective efficacy of carbon dioxide when combined with hypoxia. The review summarizes the possible influence of intermittent hypoxia and hypercapnia on aging processes in the nervous system. We considered the perspective variants of the application of hypercapnic-hypoxic influences for achieving active longevity and the prospects for the possibilities of developing hypercapnic-hypoxic training methods.
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; (Y.K.K.)
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, RUDN University, 117198 Moscow, Russia
| | - Yulia K. Komleva
- Brain Science Institute, Research Center of Neurology, 125367 Moscow, Russia; (Y.K.K.)
| | - Vladimir P. Kulikov
- Department of Ultrasound and Functional Diagnostics, Altay State Medical University, 656040 Barnaul, Russia
| | - Pavel A. Chekulaev
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | | | - Larisa D. Maltseva
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Zaripat Sh. Manasova
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Inga A. Popova
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Natalia S. Andriutsa
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Natalia V. Samburova
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Alla B. Salmina
- Brain Science Institute, Research Center of Neurology, 125367 Moscow, Russia; (Y.K.K.)
| | - Peter F. Litvitskiy
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| |
Collapse
|
2
|
Cao W, Zeng Y, Su Y, Gong H, He J, Liu Y, Li C. The involvement of oxidative stress and the TLR4/NF-κB/NLRP3 pathway in acute lung injury induced by high-altitude hypoxia. Immunobiology 2024; 229:152809. [PMID: 38788361 DOI: 10.1016/j.imbio.2024.152809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 04/13/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024]
Abstract
OBJECTIVE This study investigated the effect of oxidative stress and the TLR4/NF-κB/NLRP3 pathway on the pathogenesis of acute lung injury (ALI) induced by high-altitude hypoxia. METHODS Rats were placed in an animal hyperbaric oxygen chamber to establish a rat model of ALI induced by high-altitude hypoxia after treatment with N-acetylcysteine (NAC; a reactive oxygen species [ROS] inhibitor) or/and MCC950 (an NLPR3 inflammasome inhibitor). After modeling, the wet-to-dry weight ratio (W/D) of rat lung tissues was calculated. In lung tissues, ROS levels were detected with immunofluorescence, the enzyme activity was tested with the kit, and the expression of TLR4/NF-κB/NLRP3 pathway-related genes and proteins was measured with western blotting and qRT-PCR. The levels of inflammatory factors in the serum were quantified with ELISA. RESULTS After modeling, rats showed significantly increased W/D, ROS levels, and Malondialdehyde (MDA) concentrations and markedly diminished Superoxide dismutase (SOD) and Glutathione (GSH) concentrations in lung tissues (all P < 0.01), accompanied by substantially enhanced serum levels of TNF-α, IL-6, and IL-1β, significantly reduced serum levels of IL-10, and remarkably augmented TLR4, NLRP3, p-NF-κB p65, NF-κB p65 mRNA, and Caspase-1 expression in lung tissues (all P < 0.01). Furthermore, treatment with NAC or MCC950 alone or in combination prominently lowered the W/D of lung tissues (P < 0.01), serum levels of TNF-α (P < 0.05), IL-6 (P < 0.05), and IL-1β (P < 0.01), and NF-κB p65 expression and phosphorylation (P < 0.05, P < 0.01) while significantly increasing SOD and GSH concentrations (P < 0.05, P < 0.01) and serum levels of IL-10 (P < 0.01) in modeled rats. Meanwhile, treatment of NAC alone or combined with MCC950 significantly reduced MDA concentration and ROS levels (P < 0.05, P < 0.01) in modeled rats, and treatment of MCC950 alone or combined with NAC considerably declined TLR4, NLRP3, and Caspase-1 expression in modeled rats (P < 0.05, P < 0.01). CONCLUSION Inhibition of oxidative stress and the TLR4/NF-κB/NLRP3 pathway can ameliorate ALI in rats exposed to high-altitude hypoxia.
Collapse
Affiliation(s)
- Wangjie Cao
- School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China; Gansu Provincial Key Laboratory of Molecular Medicine and Traditional Chinese Medicine for Major Diseases, Lanzhou 730000, China; Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Lanzhou 730000, China
| | - Yuanding Zeng
- School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China; Gansu Provincial Key Laboratory of Molecular Medicine and Traditional Chinese Medicine for Major Diseases, Lanzhou 730000, China; Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Lanzhou 730000, China
| | - Yun Su
- School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China; Gansu Provincial Key Laboratory of Molecular Medicine and Traditional Chinese Medicine for Major Diseases, Lanzhou 730000, China; Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Lanzhou 730000, China.
| | - Hongxia Gong
- School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China; Gansu Provincial Key Laboratory of Molecular Medicine and Traditional Chinese Medicine for Major Diseases, Lanzhou 730000, China; Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Lanzhou 730000, China
| | - Jianzheng He
- School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China; Gansu Provincial Key Laboratory of Molecular Medicine and Traditional Chinese Medicine for Major Diseases, Lanzhou 730000, China; Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Lanzhou 730000, China
| | - Yongqi Liu
- School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China; Gansu Provincial Key Laboratory of Molecular Medicine and Traditional Chinese Medicine for Major Diseases, Lanzhou 730000, China; Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Lanzhou 730000, China
| | - Congyi Li
- School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China; Gansu Provincial Key Laboratory of Molecular Medicine and Traditional Chinese Medicine for Major Diseases, Lanzhou 730000, China; Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Lanzhou 730000, China
| |
Collapse
|
3
|
Yang WP, Li MQ, Ding J, Li JY, Wu G, Liu B, Gao YQ, Wang GH, Luo QQ. High-altitude hypoxia exposure inhibits erythrophagocytosis by inducing macrophage ferroptosis in the spleen. eLife 2024; 12:RP87496. [PMID: 38629942 PMCID: PMC11023697 DOI: 10.7554/elife.87496] [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] [Indexed: 04/19/2024] Open
Abstract
High-altitude polycythemia (HAPC) affects individuals living at high altitudes, characterized by increased red blood cells (RBCs) production in response to hypoxic conditions. The exact mechanisms behind HAPC are not fully understood. We utilized a mouse model exposed to hypobaric hypoxia (HH), replicating the environmental conditions experienced at 6000 m above sea level, coupled with in vitro analysis of primary splenic macrophages under 1% O2 to investigate these mechanisms. Our findings indicate that HH significantly boosts erythropoiesis, leading to erythrocytosis and splenic changes, including initial contraction to splenomegaly over 14 days. A notable decrease in red pulp macrophages (RPMs) in the spleen, essential for RBCs processing, was observed, correlating with increased iron release and signs of ferroptosis. Prolonged exposure to hypoxia further exacerbated these effects, mirrored in human peripheral blood mononuclear cells. Single-cell sequencing showed a marked reduction in macrophage populations, affecting the spleen's ability to clear RBCs and contributing to splenomegaly. Our findings suggest splenic ferroptosis contributes to decreased RPMs, affecting erythrophagocytosis and potentially fostering continuous RBCs production in HAPC. These insights could guide the development of targeted therapies for HAPC, emphasizing the importance of splenic macrophages in disease pathology.
Collapse
Affiliation(s)
- Wan-ping Yang
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong UniversityNantongChina
| | - Mei-qi Li
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong UniversityNantongChina
| | - Jie Ding
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong UniversityNantongChina
| | - Jia-yan Li
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong UniversityNantongChina
| | - Gang Wu
- College of High-Altitude Military Medicine, Institute of Medicine and Hygienic Equipment for High Altitude Region, Army Medical UniversityChongqingChina
- Key Laboratory of Extreme Environmental Medicine and High-Altitude Medicine, Ministry of Education of ChinaChongqingChina
| | - Bao Liu
- College of High-Altitude Military Medicine, Institute of Medicine and Hygienic Equipment for High Altitude Region, Army Medical UniversityChongqingChina
- Key Laboratory of Extreme Environmental Medicine and High-Altitude Medicine, Ministry of Education of ChinaChongqingChina
| | - Yu-qi Gao
- College of High-Altitude Military Medicine, Institute of Medicine and Hygienic Equipment for High Altitude Region, Army Medical UniversityChongqingChina
- Key Laboratory of Extreme Environmental Medicine and High-Altitude Medicine, Ministry of Education of ChinaChongqingChina
| | - Guo-hua Wang
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong UniversityNantongChina
- Department of Neurosurgery, Southwest Hospital, Army Medical UniversityChongqingChina
| | - Qian-qian Luo
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong UniversityNantongChina
| |
Collapse
|
4
|
Zhao S, Jia N, Shen Z, Pei C, Huang D, Liu J, Wang Y, Shi S, Wang X, Wang M, He Y, Wang Z. Pretreatment with Notoginsenoside R1 attenuates high-altitude hypoxia-induced cardiac injury via activation of the ERK1/2-P90RSK-Bad signaling pathway in rats. Phytother Res 2023; 37:4522-4539. [PMID: 37313866 DOI: 10.1002/ptr.7923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/19/2023] [Accepted: 05/30/2023] [Indexed: 06/15/2023]
Abstract
High-altitude cardiac injury (HACI) is one of the common tissue injuries caused by high-altitude hypoxia that may be life threatening. Notoginsenoside R1 (NG-R1), a major saponin of Panax notoginseng, exerts anti-oxidative, anti-inflammatory, and anti-apoptosis effects, protecting the myocardium from hypoxic injury. This study aimed to investigate the protective effect and molecular mechanism of NG-R1 against HACI. We simulated a 6000 m environment for 48 h in a hypobaric chamber to create a HACI rat model. Rats were pretreated with NG-R1 (50, 100 mg/kg) or dexamethasone (4 mg/kg) for 3 days and then placed in the chamber for 48 h. The effect of NG-R1 was evaluated by changes in Electrocardiogram parameters, histopathology, cardiac biomarkers, oxidative stress and inflammatory indicators, key protein expression, and immunofluorescence. U0126 was used to verify whether the anti-apoptotic effect of NG-R1 was related to the activation of ERK pathway. Pretreatment with NG-R1 can improve abnormal cardiac electrical conduction and alleviate high-altitude-induced tachycardia. Similar to dexamethasone, NG-R1 can improve pathological damage, reduce the levels of cardiac injury biomarkers, oxidative stress, and inflammatory indicators, and down-regulate the expression of hypoxia-related proteins HIF-1α and VEGF. In addition, NG-R1 reduced cardiomyocyte apoptosis by down-regulating the expression of apoptotic proteins Bax, cleaved caspase 3, cleaved caspase 9, and cleaved PARP1 and up-regulating the expression of anti-apoptotic protein Bcl-2 through activating the ERK1/2-P90RSK-Bad pathway. In conclusion, NG-R1 prevented HACI and suppressed apoptosis via activation of the ERK1/2-P90RSK-Bad pathway, indicating that NG-R1 has therapeutic potential to treat HACI.
Collapse
Affiliation(s)
- Sijing Zhao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- School of Traditional Chinese Medicine, Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Nan Jia
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zherui Shen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Caixia Pei
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Demei Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Junling Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yilan Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shihua Shi
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiaomin Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Mingjie Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yacong He
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zhenxing Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Altered Lung Heat Shock Protein-70 Expression and Severity of Sepsis-Induced Acute Lung Injury in a Chronic Kidney Disease Rat Model. Int J Mol Sci 2023; 24:ijms24065641. [PMID: 36982713 PMCID: PMC10053314 DOI: 10.3390/ijms24065641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/05/2023] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Enhanced heat shock protein-70 (HSP-70) expression in the lungs is associated with attenuated acute lung injury (ALI) in a sepsis model. Chronic kidney disease (CKD) significantly contributes to the poor prognosis of patients with sepsis. This study examined the relationship between sepsis-induced ALI severity and altered lung HSP-70 expression in CKD. Experimental rats underwent a sham operation (control group) or 5/6 nephrectomy (CKD group). Sepsis was induced with cecal ligation and puncture (CLP). Laboratory tests and lung harvest were performed in the control group (without CLP and after 3, 12, 24, and 72 h of CLP) and in the CKD group (without CLP and after 72 h of CLP). ALI was the most severe after 12 h of sepsis. The mean lung injury score at 72 h after sepsis was significantly higher in the CKD group than in the control group (4.38 versus 3.30, p < 0.01). Nonetheless, enhanced lung HSP-70 expression was not observed in the CKD group. This study shows that altered lung HSP-70 expression is associated with the worsening of sepsis-induced ALI in patients with CKD. Enhancing lung HSP-70 is a novel treatment target for patients with CKD and sepsis-induced ALI.
Collapse
|
6
|
Inflammation in Pulmonary Hypertension and Edema Induced by Hypobaric Hypoxia Exposure. Int J Mol Sci 2022; 23:ijms232012656. [PMID: 36293512 PMCID: PMC9604159 DOI: 10.3390/ijms232012656] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 11/06/2022] Open
Abstract
Exposure to high altitudes generates a decrease in the partial pressure of oxygen, triggering a hypobaric hypoxic condition. This condition produces pathophysiologic alterations in an organism. In the lung, one of the principal responses to hypoxia is the development of hypoxic pulmonary vasoconstriction (HPV), which improves gas exchange. However, when HPV is exacerbated, it induces high-altitude pulmonary hypertension (HAPH). Another important illness in hypobaric hypoxia is high-altitude pulmonary edema (HAPE), which occurs under acute exposure. Several studies have shown that inflammatory processes are activated in high-altitude illnesses, highlighting the importance of the crosstalk between hypoxia and inflammation. The aim of this review is to determine the inflammatory pathways involved in hypobaric hypoxia, to investigate the key role of inflammation in lung pathologies, such as HAPH and HAPE, and to summarize different anti-inflammatory treatment approaches for these high-altitude illnesses. In conclusion, both HAPE and HAPH show an increase in inflammatory cell infiltration (macrophages and neutrophils), cytokine levels (IL-6, TNF-α and IL-1β), chemokine levels (MCP-1), and cell adhesion molecule levels (ICAM-1 and VCAM-1), and anti-inflammatory treatments (decreasing all inflammatory components mentioned above) seem to be promising mitigation strategies for treating lung pathologies associated with high-altitude exposure.
Collapse
|
7
|
Passos FRS, Araújo-Filho HG, Monteiro BS, Shanmugam S, Araújo AADS, Almeida JRGDS, Thangaraj P, Júnior LJQ, Quintans JDSS. Anti-inflammatory and modulatory effects of steroidal saponins and sapogenins on cytokines: A review of pre-clinical research. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 96:153842. [PMID: 34952766 DOI: 10.1016/j.phymed.2021.153842] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 10/23/2021] [Accepted: 10/28/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Saponins are glycosides which, after acid hydrolysis, liberate sugar(s) and an aglycone (sapogenin) which can be triterpenoid or steroidal in nature. Steroidal saponins and sapogenins have attracted significant attention as important natural anti-inflammatory compounds capable of acting on the activity of several inflammatory cytokines in various inflammatory models. PURPOSE The aim of this review is to collect preclinical in vivo studies on the anti-inflammatory activity of steroidal saponins through the modulation of inflammatory cytokines. STUDY DESIGN AND METHODS This review was carried out through a specialized search in three databases, that were accessed between September and October, 2021, and the publication period of the articles was not limited. Information about the name of the steroidal saponins, the animals used, the dose and route of administration, the model of pain or inflammation used, the tissue and experimental method used in the measurement of the cytokines, and the results observed on the levels of cytokines was retrieved. RESULTS Forty-five (45) articles met the inclusion criteria, involving the saponins cantalasaponin-1, α-chaconine, dioscin, DT-13, lycoperoside H, protodioscin, α-solanine, timosaponin AIII and BII, trillin, and the sapogenins diosgenin, hecogenin, and ruscogenin. The surveys were carried out in seven different countries and only articles between 2007 and 2021 were found. The studies included in the review showed that the saponins and sapogenins were anti-inflammatory, antinociceptive and antioxidant and they modulate inflammatory cytokines mainly through the Nf-κB, TLR4 and MAPKs pathways. CONCLUSION Steroidal saponins and sapogenins are promising compounds in handling of pain and inflammation for the development of natural product-derived drugs. However, it is necessary to increase the methodological quality of preclinical studies, mainly blinding and sample size calculation.
Collapse
Affiliation(s)
- Fabiolla Rocha Santos Passos
- Multiuser Health Center Facility (CMulti-Saúde), Federal University of Sergipe, São Cristóvão, SE, Brazil; Health Sciences Graduate Program (PPGCS), Federal University of Sergipe, São Cristóvão, SE, Brazil; Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Marechal Rondon Avenue, S/N, Rosa Elza, São Cristóvão, Brazil
| | - Heitor Gomes Araújo-Filho
- Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Marechal Rondon Avenue, S/N, Rosa Elza, São Cristóvão, Brazil
| | - Brenda Souza Monteiro
- Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Marechal Rondon Avenue, S/N, Rosa Elza, São Cristóvão, Brazil
| | - Saravanan Shanmugam
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | | | | | | | - Lucindo José Quintans Júnior
- Multiuser Health Center Facility (CMulti-Saúde), Federal University of Sergipe, São Cristóvão, SE, Brazil; Health Sciences Graduate Program (PPGCS), Federal University of Sergipe, São Cristóvão, SE, Brazil; Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Marechal Rondon Avenue, S/N, Rosa Elza, São Cristóvão, Brazil
| | - Jullyana de Souza Siqueira Quintans
- Multiuser Health Center Facility (CMulti-Saúde), Federal University of Sergipe, São Cristóvão, SE, Brazil; Health Sciences Graduate Program (PPGCS), Federal University of Sergipe, São Cristóvão, SE, Brazil; Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Marechal Rondon Avenue, S/N, Rosa Elza, São Cristóvão, Brazil.
| |
Collapse
|
8
|
Wan Y, Huang L, Liu Y, Ji W, Li C, Ge RL. Preconditioning With Intermittent Hypobaric Hypoxia Attenuates Stroke Damage and Modulates Endocytosis in Residual Neurons. Front Neurol 2022; 12:750908. [PMID: 34975719 PMCID: PMC8715922 DOI: 10.3389/fneur.2021.750908] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 11/26/2021] [Indexed: 01/01/2023] Open
Abstract
Background: Moderate hypobaric hypoxia induces cerebral ischemic tolerance. We investigated the optimal method for applying hypobaric hypoxia preconditioning at 5,000 m to ischemic brain tissue and combined it with proteomics to determine the mechanisms underlying this effect. Methods: Male SD rats were randomly grouped as S (sham, n = 20), M (middle cerebral artery occlusion [MCAO], n = 28), H2M (intermittent hypobaric hypoxia preconditioned MCAO group, 2 h/day, 10 days, n = 20), H6M (intermittent hypobaric hypoxia preconditioned MCAO group, 6 h/day, 10 days, n = 28), and HpM (persistent hypobaric hypoxia preconditioned MCAO group, 10 days, n = 28). The permanent MCAO model was established based on the Zea Longa method. Infarction was assessed with the modified neurological severity score (mNSS) and 2,3,5-triphenyl tetrazolium chloride staining. The total protein expression of the neuron-specific nuclear protein (NeuN), cysteinyl aspartate specific proteinase 3 (caspase-3), cleaved-caspase-3, and interleukin 6 (IL-6) was determined using western blotting. We assessed the peri-infarct cortex's ultrastructural changes. A label-free proteomic study and western blot verification were performed on the most effective preconditioned group. Results: The H6M group showed a lower infarct volume (p = 0.0005), lower mNSS score (p = 0.0009) than the M group. The H2M showed a lower level of IL-6 (p = 0.0213) than the M group. The caspase-3 level decreased in the H2M (p = 0.0002), H6M (p = 0.0025), and HpM groups (p = 0.0054) compared with that in the M group. Cleaved-caspase-3 expression decreased in the H2M (p = 0.0011), H6M (p < 0.0001), and HpM groups (p < 0.0001) compared with that in the M group. The neurons' ultrastructure and the blood-brain barrier in the peri-infarct tissue improved in the H2M and H6M groups. Immunofluorescence revealed increased NeuN-positive cells in the peri-infarct tissue in the H6M group (p = 0.0003, H6M vs. M). Protein expression of Chmp1a, Arpc5, and Hspa2 factors related to endocytosis were upregulated in the H6M compared with those of the M group (p < 0.05 for all) on western blot verification of label-free proteomics. Conclusions: Intermittent hypobaric hypoxia preconditioning exerts a neuroprotective effect in a rat stroke model. Persistent hypobaric hypoxia stimulation exhibited no significant neuroprotective effect. Intermittent hypoxic preconditioning for 6 h/day for 10 days upregulates key proteins in clathrin-dependent endocytosis of neurons in the cortex.
Collapse
Affiliation(s)
- Yaqi Wan
- Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province, Research Center for High Altitude Medicine, Qinghai University, Xining, China.,Qinghai Provincial People's Hospital, Xining, China
| | - Lu Huang
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yanmin Liu
- Qinghai Provincial People's Hospital, Xining, China
| | - Weizhong Ji
- Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province, Research Center for High Altitude Medicine, Qinghai University, Xining, China.,Qinghai Provincial People's Hospital, Xining, China
| | - Changxing Li
- Department of Basic Medicine, Qinghai University, Xining, China
| | - Ri-Li Ge
- Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province, Research Center for High Altitude Medicine, Qinghai University, Xining, China
| |
Collapse
|
9
|
Alsharif KF, Almalki AA, Alsanie WF, Alzahrani KJ, Kabrah SM, Elshopakey GE, Alghamdi AAA, Lokman MS, Sberi HA, Bauomy AA, Albrakati A, Ramadan SS, Kassab RB, Abdel Moneim AE, Salem FEH. Protocatechuic acid attenuates lipopolysaccharide-induced septic lung injury in mice: The possible role through suppressing oxidative stress, inflammation and apoptosis. J Food Biochem 2021; 45:e13915. [PMID: 34472624 DOI: 10.1111/jfbc.13915] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 07/14/2021] [Accepted: 08/16/2021] [Indexed: 12/11/2022]
Abstract
Here, we investigated the protective efficacy of protocatechuic acid (PCA) against lipopolysaccharide (LPS)-induced septic lung injury. Eighty-two male Balb/c mice were divided into six groups: control, PCA30 (30 mg/kg), LPS (10 mg/kg), PCA10-LPS, PCA20-LPS, and PCA30-LPS treated with 10, 20 and 30 mg/kg PCA, respectively, for seven days before intraperitoneal LPS injection. PCA pre-treatment, especially at higher dose, significantly reduced LPS-induced lung tissue injury as indicated by increased heat shock protein 70 and antioxidant molecules (reduced glutathione, superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase) accompanied by lower oxidative stress indices (malondialdehyde and nitric oxide). PCA administration decreased inflammatory mediators including myeloperoxidase, nuclear factor kappa B (NF-κB p65), and pro-inflammatory cytokines, and prevented the development of apoptotic events in the lung tissue. At the molecular level, PCA downregulated mRNA expression of nitric oxide synthase 2, C/EBP homologous protein, and high mobility group box1 in the lungs of all PCA-LPS treated mice. Thus, PCA-pre-treatment effectively counteracted sepsis-induced acute lung injury in vivo by promoting and antioxidant status, while inhibiting inflammation and apoptosis. PRACTICAL IMPLICATIONS: Sepsis-mediated organ dysfunction and high mortality is aggravated by acute lung injury (ALI). Therefore, new therapeutic approaches are needed to encounter sepsis-mediated ALI. Protocatechuic acid (PCA) is a naturally occurring phenolic acid with various biological and pharmacological activities. PCA is abundant in edible plants including Allium cepa L., Oryza sativa L., Hibiscus sabdariffa, Prunus domestica L., and Eucommia ulmoides. In this investigation we studied the potential protective role of pure PCA (10, 20 and 30 mg/kg) on LPS-mediated septic lung injury in mice through examining oxidative challenge, inflammatory response, apoptotic events and histopathological changes in addition to evaluating the levels and mRNA expression of heat shock protein 70, C/EBP homologous protein and high mobility group box1 in the lung tissue. The recorded results showed that PCA pre-administration was able to significantly abrogate the damages in the lung tissue associated septic response. This protective effect comes from its strong antioxidant, anti-inflammatory, and anti-apoptotic activities, suggesting that PCA may be applied to alleviate ALI associated with the development of sepsis.
Collapse
Affiliation(s)
- Khalaf F Alsharif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Abdulraheem Ali Almalki
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Walaa F Alsanie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Khalid J Alzahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Saeed M Kabrah
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm AlQura University, Mecca, Saudi Arabia
| | - Gehad E Elshopakey
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | | | - Maha S Lokman
- Biology Department, College of Science and Humanities, Prince Sattam Bin Abdul Aziz University, Alkharj, Saudi Arabia.,Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Hassan Al Sberi
- Basic Medical Science, Histopathology Department, National Organization for Drug Control and Research, Giza, Egypt.,Department of Biology, Faculty of Science, Taif University, Taif, Saudi Arabia
| | - Amira A Bauomy
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt.,Department of Science Laboratories, College of Science and Arts, Qassim University, ArRass, Saudi Arabia
| | - Ashraf Albrakati
- Department of Human Anatomy, College of Medicine, Taif University, Taif, Saudi Arabia
| | - Shimaa S Ramadan
- Department of Chemistry, Faculty of Science, Helwan University, Cairo, Egypt
| | - Rami B Kassab
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt.,Department of Biology, Faculty of Science and Arts, Al Baha University, Almakhwah, Saudi Arabia
| | - Ahmed E Abdel Moneim
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Fatma Elzahraa H Salem
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| |
Collapse
|
10
|
Scultetus AH, Jefferson MA, Haque A, Hubbell JN, Arnaud FG, Moon-Massat P, McCarron RM, Malone DL. Histopathological Evidence of Multiple Organ Damage After Simulated Aeromedical Evacuation in a Swine Acute Lung Injury Model. Mil Med 2020; 185:57-66. [PMID: 32074309 DOI: 10.1093/milmed/usz248] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
INTRODUCTION Rapid aeromedical evacuation (AE) is standard of care in current conflicts. However, not much is known about possible effects of hypobaric conditions. We investigated possible effects of hypobaria on organ damage in a swine model of acute lung injury. METHODS Lung injury was induced in anesthetized swine via intravenous oleic acid infusion. After a stabilization phase, animals were subjected to a 4 hour simulated AE at 8000 feet (HYPO). Control animals were kept at normobaria. After euthanasia and necropsy, organ damage was assessed by combined scores for hemorrhage, inflammation, edema, necrosis, and microatelectasis. RESULTS Hemodynamic, neurological, or hematologic measurements were similar prior to transport. Hemodynamic instability became apparent during the last 2 hours of transport in the HYPO group. Histological injury scores in the HYPO group were higher for all organs (lung, kidney, liver, pancreas, and adrenal glands) except the brain, with the largest difference in the lungs (P < 0.001). CONCLUSIONS Swine with mild acute lung injury subjected to a 4 hour simulated AE showed more injury to most organs and, in particular, to the lungs compared with ground transport. This may exacerbate otherwise subclinical pathology and, eventually, manifest as abnormalities in gas exchange or possibly end-organ function.
Collapse
Affiliation(s)
- Anke H Scultetus
- Neuro Trauma Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD 20910.,Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814
| | - Michelle A Jefferson
- Department of Pathology, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910
| | - Ashraful Haque
- Neuro Trauma Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD 20910.,Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817
| | - Jordan N Hubbell
- Neuro Trauma Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD 20910.,Parsons, 100 West Walnut Street, Pasadena, CA 91124.,Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817
| | - Francoise G Arnaud
- Neuro Trauma Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD 20910.,Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814.,Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817
| | - Paula Moon-Massat
- Neuro Trauma Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD 20910.,Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817
| | - Richard M McCarron
- Neuro Trauma Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD 20910.,Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814
| | - Debra L Malone
- Neuro Trauma Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD 20910.,Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814.,Department of Surgery, Walter Reed National Military Medical Center, 4494 North Palmer Road, Bethesda, MD 20889
| |
Collapse
|
11
|
Chao CM, Chen CL, Niu KC, Lin CH, Tang LY, Lin LS, Chang CP. Hypobaric hypoxia preconditioning protects against hypothalamic neuron apoptosis in heat-exposed rats by reversing hypothalamic overexpression of matrix metalloproteinase-9 and ischemia. Int J Med Sci 2020; 17:2622-2634. [PMID: 33162790 PMCID: PMC7645337 DOI: 10.7150/ijms.47560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/24/2020] [Indexed: 12/03/2022] Open
Abstract
Background: Hypoxia-inducible factor-1α (HIF-1α), heat shock protein-72 (HSP-72), hemeoxygenase-1 (HO-1), and matrix metalloproteinase-9 (MMP-9) have been identified as potential therapeutic targets in the brain for cerebral ischemia. To elucidate their underlying mechanisms, we first aimed to ascertain whether these proteins participate in the pathogenesis of heat-induced ischemic damage to the hypothalamus of rats. Second, we investigated whether hypobaric hypoxia preconditioning (HHP) attenuates heat-induced hypothalamic ischemic/hypoxic injury by modulating these proteins in situ. Methods: Anesthetized rats treated with or without HHP were subjected to heat stress. Hypothalamic ischemic/hypoxic damage was evaluated by measuring hypothalamic levels of cerebral blood flow (CBF), partial oxygen pressure (PO2), and hypothalamic temperature via an implanted probe. Hypothalamic apoptotic neurons were counted by measuring the number of NeuN/caspase-3/DAPI triple-stained cells. Hypothalamic protein expression of HIF-1α, HSP-72, HO-1, and MMP-9 was determined biochemically. Results: Before the start of the thermal experiments, rats were subjected to 5 hours of HHP (0.66 ATA or 18.3% O2) daily for 5 consecutive days per week for 2 weeks, which led to significant loss of body weight, reduced brown adipose tissue (BAT) wet weight and decreased body temperature. The animals were then subjected to thermal studies. Twenty minutes after heat stress, heat-exposed rats not treated with HHP displayed significantly higher core and hypothalamic temperatures, hypothalamic MMP-9 levels, and numbers of hypothalamic apoptotic neurons but significantly lower mean blood pressure, hypothalamic blood flow, and PO2 values than control rats not exposed to heat. In heat-exposed rats, HHP significantly increased the hypothalamic levels of HIF-1α, HSP-72, and HO-1 but significantly alleviated body and hypothalamic hyperthermia, hypotension, hypothalamic ischemia, hypoxia, neuronal apoptosis and degeneration. Conclusions: HHP may protect against hypothalamic ischemic/hypoxic injury and overexpression of MMP-9 by upregulating the hypothalamic expression of HIF-1α, HSP-72, and HO-1 in rats subjected to heatstroke.
Collapse
Affiliation(s)
- Chien-Ming Chao
- Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan
- Department of Nursing, Min-Hwei College of Health Care Management, Tainan, Taiwan
| | - Chun-Liang Chen
- Department of Gastroenterology and General Surgery, Chi Mei Medical Hospital, Chiali, Tainan, Taiwan
| | - Ko-Chi Niu
- Department of Hyperbaric Oxygen, Chi Mei Medical Center, Tainan, Taiwan
| | - Cheng-Hsien Lin
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Ling-Yu Tang
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Lieh-Sheng Lin
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Ching-Ping Chang
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| |
Collapse
|
12
|
Nimje MA, Patir H, Tirpude RK, Reddy PK, Kumar B. Physiological and oxidative stress responses to intermittent hypoxia training in Sprague Dawley rats. Exp Lung Res 2020; 46:376-392. [PMID: 32930002 DOI: 10.1080/01902148.2020.1821263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
AIM Rapid ascent to high altitude and inability to acclimatize lead to high-altitude illnesses. Intermittent hypoxia (IH) conditioning has been hypothesized as a non-pharmacological strategy aiming to improve adaptive responses during high altitude ascent. In the recent years, IH training (IHT) has become increasingly popular among recreational and professional athletes owing to its ability to mitigate high altitude related problems. This study aimed at exploring the role of IHT in altitude acclimatization. METHODS Male Sprague Dawley rats were subjected to IHT for 4 h consecutively for 5 days at 12% FiO2 under normobaric conditions. To assess the effect of IHT in hypoxic acclimatization, animals were further exposed to extreme hypoxia (EH) at 8% FiO2. Oxygen saturation (SpO2), respiratory rate and heart rate were recorded during the exposure. Oxidative stress (ROS, MDA, and 4-HNE) and histopathological examinations were studied in the lung tissue sections. Hypoxia biomarkers, HIF-1α, EPO, VEGF, and BPGM were evaluated through western blotting in the lung tissue. RESULTS Assessment of the IHT showed that SpO2 levels were found to be higher in the IH trained rats with a statistical difference of p < 0.01 in the first hour of hypoxia exposure as compared to the untrained rats. There was a significantly higher (p < 0.001) generation of ROS and MDA in the untrained rats as compared to the trained rats. Lipid peroxidation markers and systemic inflammatory marker were found to be expressed at much higher level in the untrained rats. There was a higher expression of HIF-1α (1.24-fold ↑), VEGF (1.14-fold ↑) and decrease in EPO (1.43-fold ↓) in the untrained rats as compared to trained rats. CONCLUSIONS Preconditioning with IHT resulted in the reduction in hypoxia induced oxidative stress during extreme hypoxia exposure and thus, maintaining redox balance as well as adjustment in the physiological changes in rats.
Collapse
Affiliation(s)
- Megha A Nimje
- Defence Institute of Physiology and Allied Sciences (DIPAS), (DRDO), Timarpur, Delhi, India
| | - Himadri Patir
- Defence Institute of Physiology and Allied Sciences (DIPAS), (DRDO), Timarpur, Delhi, India
| | - Rajesh Kumar Tirpude
- Defence Institute of Physiology and Allied Sciences (DIPAS), (DRDO), Timarpur, Delhi, India
| | - Prasanna K Reddy
- Defence Institute of Physiology and Allied Sciences (DIPAS), (DRDO), Timarpur, Delhi, India
| | - Bhuvnesh Kumar
- Defence Institute of Physiology and Allied Sciences (DIPAS), (DRDO), Timarpur, Delhi, India
| |
Collapse
|
13
|
Tregub PP, Malinovskaya NA, Morgun AV, Osipova ED, Kulikov VP, Kuzovkov DA, Kovzelev PD. Hypercapnia potentiates HIF-1α activation in the brain of rats exposed to intermittent hypoxia. Respir Physiol Neurobiol 2020; 278:103442. [DOI: 10.1016/j.resp.2020.103442] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/19/2020] [Accepted: 04/06/2020] [Indexed: 12/30/2022]
|
14
|
Xie Y, Qin S, Zhang R, Wu H, Sun G, Liu L, Hou X. The Effects of High-Altitude Environment on Brain Function in a Seizure Model of Young-Aged Rats. Front Pediatr 2020; 8:561. [PMID: 33072659 PMCID: PMC7534851 DOI: 10.3389/fped.2020.00561] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/03/2020] [Indexed: 12/25/2022] Open
Abstract
In this study, we examined the effects of high-altitude environment on the brain function of a young-rat seizure model. Two-hundred healthy, 3-week old, male rats were selected and equally divided into the plateau and plain groups. The plateau group was preconditioned in a simulated 5,000-m altitude (barometric pressure [PB], 405 mmHg; partial pressure of oxygen [PO2], 84 mmHg) for 6 h/day for 7 days, while the plain group was kept in the ordinary atmospheric environment (PB, 760 mmHg; PO2, 157 mmHg) for 7 days. After preconditioning, rats were administered pentylenetetrazol (PTZ) to generate level-4 or stronger seizures. Electroencephalogram (EEG) signals were recorded (16 rats/group); the histology and apoptosis of hippocampal tissue were evaluated (6 rats/group); and spatial learning and memory were examined in the Morris water maze (12 rats/group; 6-weeks old). To induce a level 4 or stronger seizure successfully, a significantly higher PTZ dose was used in the plateau (81.32 ± 21.57 mg/kg) than in the plain group (63.41 ± 19.77 mg/kg, p < 0.01); however, the plateau group survival rate was significantly lower than that of the plain group (26.2 vs. 42.9%, p < 0.05). EEG parameters did not differ between the two groups. Histological analysis revealed that in the plateau group, more neurons were observed (p < 0.001), especially in DG and CA1 areas, and less apoptotic cells were found in DG areas (p = 0.035), comparing with the plain group. No differences were found between the two groups in any of the parameters examined in the Morris water maze. Our results show that the disease outcome caused by low pressure and low oxygen environment in the plateau group was different to that in the plain group. The high drug dosage to induce seizures in the plateau group, accompanied by increased mortality rates after seizures, indicates that the seizure threshold may be higher in the plateau than in the plain group. Moreover, based on the histological findings, the plateau environment seems to exert a protective effect on brain development after seizures only for survived individuals with mild conditions.
Collapse
Affiliation(s)
- Yao Xie
- Pediatric Department, Peking University First Hospital, Beijing, China
| | - Shenglan Qin
- Pediatric Department, People's Hospital of Tibet Autonomous Region, Tibet, China
| | - Rui Zhang
- Pediatric Department, Peking University First Hospital, Beijing, China
| | - Hong Wu
- Pediatric Department, People's Hospital of Tibet Autonomous Region, Tibet, China
| | - Guoyu Sun
- Pediatric Department, Peking University First Hospital, Beijing, China
| | - Lili Liu
- Pediatric Department, Peking University First Hospital, Beijing, China
| | - Xinlin Hou
- Pediatric Department, Peking University First Hospital, Beijing, China
| |
Collapse
|
15
|
Hypercapnic hypoxia as a potential means to extend life expectancy and improve physiological activity in mice. Biogerontology 2019; 20:677-686. [DOI: 10.1007/s10522-019-09821-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/10/2019] [Indexed: 01/09/2023]
|
16
|
Wu G, Xu G, Chen DW, Gao WX, Xiong JQ, Shen HY, Gao YQ. Hypoxia Exacerbates Inflammatory Acute Lung Injury via the Toll-Like Receptor 4 Signaling Pathway. Front Immunol 2018; 9:1667. [PMID: 30083155 PMCID: PMC6064949 DOI: 10.3389/fimmu.2018.01667] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 07/04/2018] [Indexed: 12/13/2022] Open
Abstract
Acute lung injury (ALI) is characterized by non-cardiogenic diffuse alveolar damage and often leads to a lethal consequence, particularly when hypoxia coexists. The treatment of ALI remains a challenge: pulmonary inflammation and hypoxia both contribute to its onset and progression and no effective prevention approach is available. Here, we aimed to investigate the underlying mechanism of hypoxia interaction with inflammation in ALI and to evaluate hypoxia-inducible factor 1 alpha (HIF-1α)—the crucial modulator in hypoxia—as a potential therapeutic target against ALI. First, we developed a novel ALI rat model induced by a combined low-dose of lipopolysaccharides (LPS) with acute hypoxia. Second, we used gene microarray analysis to evaluate the inflammatory profiles of bronchi alveolar lavage fluid cells of ALI rats. Third, we employed an alveolar macrophage cell line, NR8383 as an in vitro system together with a toll-like receptor 4 (TLR4) antagonist TAK-242, to verify our in vivo findings from ALI animals. Finally, we tested the therapeutic effects of HIF-1α augmentation against inflammation and hypoxia in ALI. We demonstrated that (i) LPS upregulated inflammatory genes, tumor necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1β), and interleukin-6 (IL-6), in the alveolar macrophages of ALI rats, which were further enhanced when ALI combined with hypoxia; (ii) hypoxia exposure could further enhance the upregulation of alveolar macrophageal TLR4 that was noticed in LPS-induced inflammatory ALI, conversely, TLR4 antagonist TAK-242 could suppress the macrophageal expression of TLR4 and inflammatory cytokines, including TNF-α, IL-1β, and IL-6, suggesting that the TLR4 signaling pathway as a central link between inflammation and hypoxia in ALI; (iii) manipulation of HIF-1α in vitro could suppress TLR4 expression induced by combined LPS and hypoxia, via suppressing promoter activity of the TLR4 gene; (iv) preconditioning augmentation of HIF-1α in vivo by HIF hydroxylase inhibitor, DMOG excreted protection against inflammatory, and hypoxic processes in ALI. Together, we see that hypoxia can exacerbate inflammation in ALI via the activation of the TLR4 signaling pathway in alveolar macrophages and predispose impairment of the alveolar-capillary barrier in the development of ALI. Targeting HIF-1α can suppress TLR4 expression and macrophageal inflammation, suggesting the potential therapeutic and preventative value of HIF-1α/TLR4 crosstalk pathway in ALI.
Collapse
Affiliation(s)
- Gang Wu
- College of High Altitude Military Medicine, Institute of Medicine and Hygienic Equipment for High Altitude Region, Army Medical University, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Gang Xu
- College of High Altitude Military Medicine, Institute of Medicine and Hygienic Equipment for High Altitude Region, Army Medical University, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - De-Wei Chen
- Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China.,Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
| | - Wen-Xiang Gao
- College of High Altitude Military Medicine, Institute of Medicine and Hygienic Equipment for High Altitude Region, Army Medical University, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Jian-Qiong Xiong
- Intensive Care Unit, Southwest Hospital, Army Medical University, Chongqing, China
| | - Hai-Ying Shen
- Robert Stone Dow Laboratories, Legacy Research Institute, Legacy Health, Portland, OR, United States
| | - Yu-Qi Gao
- College of High Altitude Military Medicine, Institute of Medicine and Hygienic Equipment for High Altitude Region, Army Medical University, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| |
Collapse
|
17
|
Zeng H, Yang L, Zhang X, Chen Y, Cai J. Dioscin prevents LPS‑induced acute lung injury through inhibiting the TLR4/MyD88 signaling pathway via upregulation of HSP70. Mol Med Rep 2018; 17:6752-6758. [PMID: 29512786 DOI: 10.3892/mmr.2018.8667] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 06/06/2017] [Indexed: 11/06/2022] Open
Abstract
Dioscin, as a type of important natural steroidal saponin, has widespread sources, primarily from the fenugreek plant, which is an important raw material in the production of synthetic steroid hormone drugs. Dioscin has anti‑tumor, anti‑inflammatory, antioxidant and other significant pharmacological effects with high medicinal value. The present work aimed to research the protective effect and underlying mechanisms by which dioscin prevents acute lung injury (ALI). Mice were injected with 5 mg/kg LPS to induce lung injury. Mice were treated with dioscin (20, 40 and 60 mg/kg) following LPS‑induced lung injury. Treatment with dioscin significantly decreased total number of alveolar macrophages, water content of lung and total protein concentration in ALI mice. Dioscin treatment significantly suppressed the ALI‑induced interleukin (IL)‑1B, IL‑6, tumor necrosis factor‑α, nuclear factor (NF)‑κB, myeloperoxidase, interferon‑γ and intercellular adhesion molecule‑1 activities in ALI rats. Following this, the authors identified that dioscin significantly also suppressed cyclooxygenase‑2, heat shock protein 70, Toll‑like receptor 4, MyD88 and NF‑κB protein expression in ALI rats. The results suggested that dioscin prevents LPS‑induced ALI through inhibiting the TLR4/MyD88 signaling pathway via upregulation of HSP70.
Collapse
Affiliation(s)
- Huiqing Zeng
- Department of Respiration Medicine, Zhongshan Hospital of Xiamen University, P.R. China
| | - Lijuan Yang
- Basic Medical College, Fujian Medical University, Xiamen, Fujian 361004, P.R. China
| | - Xiaobin Zhang
- Department of Respiration Medicine, Zhongshan Hospital of Xiamen University, P.R. China
| | - Yan Chen
- Department of Respiration Medicine, Zhongshan Hospital of Xiamen University, P.R. China
| | - Jianghang Cai
- Department of Respiration Medicine, Zhongshan Hospital of Xiamen University, P.R. China
| |
Collapse
|
18
|
Sheppard RL, Swift JM, Hall A, Mahon RT. The Influence of CO 2 and Exercise on Hypobaric Hypoxia Induced Pulmonary Edema in Rats. Front Physiol 2018. [PMID: 29541032 PMCID: PMC5835685 DOI: 10.3389/fphys.2018.00130] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Introduction: Individuals with a known susceptibility to high altitude pulmonary edema (HAPE) demonstrate a reduced ventilation response and increased pulmonary vasoconstriction when exposed to hypoxia. It is unknown whether reduced sensitivity to hypercapnia is correlated with increased incidence and/or severity of HAPE, and while acute exercise at altitude is known to exacerbate symptoms the effect of exercise training on HAPE susceptibility is unclear. Purpose: To determine if chronic intermittent hypercapnia and exercise increases the incidence of HAPE in rats. Methods: Male Wistar rats were randomized to sedentary (sed-air), CO2 (sed-CO2,) exercise (ex-air), or exercise + CO2 (ex-CO2) groups. CO2 (3.5%) and treadmill exercise (15 m/min, 10% grade) were conducted on a metabolic treadmill, 1 h/day for 4 weeks. Vascular reactivity to CO2 was assessed after the training period by rheoencephalography (REG). Following the training period, animals were exposed to hypobaric hypoxia (HH) equivalent to 25,000 ft for 24 h. Pulmonary injury was assessed by wet/dry weight ratio, lung vascular permeability, bronchoalveolar lavage (BAL), and histology. Results: HH increased lung wet/dry ratio (HH 5.51 ± 0.29 vs. sham 4.80 ± 0.11, P < 0.05), lung permeability (556 ± 84 u/L vs. 192 ± 29 u/L, P < 0.001), and BAL protein (221 ± 33 μg/ml vs. 114 ± 13 μg/ml, P < 0.001), white blood cell (1.16 ± 0.26 vs. 0.66 ± 0.06, P < 0.05), and platelet (16.4 ± 2.3, vs. 6.0 ± 0.5, P < 0.001) counts in comparison to normobaric normoxia. Vascular reactivity was suppressed by exercise (−53% vs. sham, P < 0.05) and exercise+CO2 (−71% vs. sham, P < 0.05). However, neither exercise nor intermittent hypercapnia altered HH-induced changes in lung wet/dry weight, BAL protein and cellular infiltration, or pulmonary histology. Conclusion: Exercise training attenuates vascular reactivity to CO2 in rats but neither exercise training nor chronic intermittent hypercapnia affect HH- induced pulmonary edema.
Collapse
Affiliation(s)
- Ryan L Sheppard
- Department of Submarine Medicine and Survival Systems Groton, Naval Submarine Medical Research Laboratory, Groton, CT, United States.,Department of Undersea Medicine, Walter Reed Army Institute of Research and Naval Medical Research Center, Silver Spring, MD, United States
| | - Joshua M Swift
- Department of Undersea Medicine, Walter Reed Army Institute of Research and Naval Medical Research Center, Silver Spring, MD, United States
| | - Aaron Hall
- Department of Undersea Medicine, Walter Reed Army Institute of Research and Naval Medical Research Center, Silver Spring, MD, United States
| | - Richard T Mahon
- Department of Undersea Medicine, Walter Reed Army Institute of Research and Naval Medical Research Center, Silver Spring, MD, United States
| |
Collapse
|
19
|
Luo H, Zhou DJ, Chen Z, Zhou QQ, Wu K, Tian K, Li ZW, Xiao ZL. Establishment and evaluation of an experimental rat model for high-altitude intestinal barrier injury. Exp Ther Med 2016; 13:475-482. [PMID: 28352318 PMCID: PMC5348649 DOI: 10.3892/etm.2016.4012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 10/28/2016] [Indexed: 01/19/2023] Open
Abstract
In the present study an experimental high-altitude intestinal barrier injury rat model was established by simulating an acute hypoxia environment, to provide an experimental basis to assess the pathogenesis, prevention and treatment of altitude sickness. A total of 70 healthy male Sprague-Dawley rats were divided into two groups: Control group (group C) and a high-altitude hypoxia group (group H). Following 2 days adaptation, the rats in group H were exposed to a simulated 4,000-m, high-altitude hypoxia environment for 3 days to establish the experimental model. To evaluate the model, bacterial translocation, serum lipopolysaccharide level, pathomorphology, ultrastructure and protein expression in rats were assessed. The results indicate that, compared with group C, the rate of bacterial translocation and the apoptotic index of intestinal epithelial cells were significantly higher in group H (P<0.01). Using a light microscope it was determined that the intestinal mucosa was thinner in group H, there were fewer epithelial cells present and the morphology was irregular. Observations with an electron microscope indicated that the intestinal epithelial cells in group H were injured, the spaces among intestinal villi were wider, the tight junctions among cells were open and lanthanum nitrate granules (from the fixing solution) had diffused into the intestinal mesenchyme. The expression of the tight junction protein occludin was also decreased in group H. Therefore, the methods applied in the present study enabled the establishment of a stable, high-altitude intestinal barrier injury model in rats.
Collapse
Affiliation(s)
- Han Luo
- Respiratory Department, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Dai-Jun Zhou
- The Fourth Institute of Field Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| | - Zhang Chen
- Respiratory Department, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Qi-Quan Zhou
- High Altitude Military Medical Science Academy, Third Military Medical University, Chongqing 400038, P.R. China
| | - Kui Wu
- Respiratory Department, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Kun Tian
- Respiratory Department, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Zhi-Wei Li
- Respiratory Department, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Zhen-Liang Xiao
- Respiratory Department, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| |
Collapse
|
20
|
Wang Q, Hu L, Hu Y, Gong G, Tan H, Deng L, Sun X, Yi X, Sun Y, Wu W, Li T. Carbon Monoxide-Saturated Hemoglobin-Based Oxygen Carriers Attenuate High-Altitude-Induced Cardiac Injury by Amelioration of the Inflammation Response and Mitochondrial Oxidative Damage. Cardiology 2016; 136:180-191. [DOI: 10.1159/000448652] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/21/2016] [Indexed: 11/19/2022]
Abstract
Objective: To investigate the therapeutic effect of carbon monoxide (CO) on high-altitude hypoxia-induced cardiac damage. Methods: Forty male C57BL/6 mice were randomly divided into 4 groups. The mice were exposed to normoxia or simulated 5,500-meter high-altitude hypoxia in a hypobaric chamber for 7 days. During the first 3 days, the mice were pretreated with CO-saturated hemoglobin (Hb)-based oxygen carrier (CO-HBOC), oxygen-saturated hemoglobin-based oxygen carrier (O2-HBOC) at a dose of 0.3 g Hb/kg/day or an equivalent volume of saline. The in vivo left ventricle function, cardiac enzyme release, histopathological changes, apoptosis and inflammation were also measured. Results: High-altitude hypoxia induced significant cardiac damage, as demonstrated by impaired cardiac function and increased proapoptotic, proinflammatory and pro-oxidant markers. Pretreatment with CO-HBOC significantly improved cardiac performance, reduced cardiac enzyme release and limited myocardial apoptosis. The increased inflammatory response was also suppressed. In addition to the preserved mitochondrial structure, hypobaric hypoxia-induced mitochondrial oxidative damage was remarkably attenuated. Moreover, these antiapoptotic and antioxidative effects were accompanied by an upregulated phosphorylation of Akt, ERK and STAT3. Conclusion: This study demonstrated that CO-HBOC provides a promising protective effect on high-altitude hypoxia-induced myocardial injury, which is mediated by the inhibition of inflammation and mitochondrial oxidative damage.
Collapse
|
21
|
Ünver R, Deveci F, Kırkıl G, Telo S, Kaman D, Kuluöztürk M. Serum Heat Shock Protein Levels and the Relationship of Heat Shock Proteins with Various Parameters in Chronic Obstructive Pulmonary Disease Patients. Turk Thorac J 2016; 17:153-159. [PMID: 29404146 PMCID: PMC5783095 DOI: 10.5152/turkthoracj.2016.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/13/2016] [Indexed: 01/11/2023]
Abstract
OBJECTIVES Chronic Obstructive Pulmonary Disease (COPD) is accompanied by increased cellular stress and inflammation. Most of the Heat Shock Proteins (HSPs) have strong cytoprotective effects. The role of HSPs in COPD pathogenesis has not determined completely. We investigated the serum level of HSPs in COPD patients, smokers without COPD and healthy non-smoking controls. Also, we evaluated the relationship of HSPs with various parameters (inflammatory, oxidative, functional status, quality of life) in COPD patients. MATERIAL AND METHODS The levels of stress protein (HSP27, HSP70, HSP60, HSP90, CyPA), interleukin-6, C-reactive protein and malondialdehyde were measured in 16 healthy non-smoker, 14 smokers without COPD and 50 patients with stable COPD. Pulmonary function tests (PFT) and arterial blood gases parameters were measured. Health Related Quality of Life was evaluated and exercise capacity was measured with 6 minute walking test. RESULTS Only HSP27 levels was significantly higher in COPD patients when compared with both healthy non-smoker and smokers without COPD (for both, p< 0.001). There was a weak-moderate negative correlation between serum levels of HSP27 and PFT parameters and between HSP27 levels and PaO2. Serum levels of HSP27 showed a weak-moderate positive correlation with symptom, activity and total scores. Subjects evaluated only smokers without COPD and patients with COPD; HSP27 had an area under the curve (AUC) in the receiver operating characteristic (ROC) curve of 0.819 (0.702-0.935; 95% CI; p= 0.000). CONCLUSION Increased serum levels of HSP27 was found in COPD patients and our results showed sensitivity and specificity of serum HSP27 as diagnostic markers for COPD.
Collapse
Affiliation(s)
- Ramazan Ünver
- Department of Chest Diseases, Fırat University Faculty of Medicine, Elazığ, Turkey
| | - Figen Deveci
- Department of Chest Diseases, Fırat University Faculty of Medicine, Elazığ, Turkey
| | - Gamze Kırkıl
- Department of Chest Diseases, Fırat University Faculty of Medicine, Elazığ, Turkey
| | - Selda Telo
- Department of Biochemestry, Fırat University Faculty of Medicine, Elazığ, Turkey
| | - Dilara Kaman
- Department of Biochemestry, Fırat University Faculty of Medicine, Elazığ, Turkey
| | - Mutlu Kuluöztürk
- Department of Chest Diseases, Fırat University Faculty of Medicine, Elazığ, Turkey
| |
Collapse
|
22
|
Parseghian MH, Hobson ST, Richieri RA. Targeted heat shock protein 72 for pulmonary cytoprotection. Ann N Y Acad Sci 2016; 1374:78-85. [PMID: 27152638 DOI: 10.1111/nyas.13059] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 02/29/2016] [Accepted: 03/09/2016] [Indexed: 12/12/2022]
Abstract
Heat shock protein 72 (HSP72) is perhaps the most important member of the HSP70 family of proteins, given that it is induced in a wide variety of tissues and cells to combat stress, particularly oxidative stress. Here, we review independent observations of the critical role this protein plays as a pulmonary cytoprotectant and discuss the merits of developing HSP72 as a therapeutic for rapid delivery to cells and tissues after a traumatic event. We also discuss the fusion of HSP72 to a cell-penetrating single-chain Fv antibody fragment derived from mAb 3E10, referred to as Fv-HSP70. This fusion construct has been validated in vivo in a cerebral infarction model and is currently in testing as a clinical therapeutic to treat ischemic events and as a fieldable medical countermeasure to treat inhalation of toxicants caused by terrorist actions or industrial accidents.
Collapse
Affiliation(s)
| | - Stephen T Hobson
- Rubicon Biotechnology, Lake Forest, California.,Seacoast Science, Inc, Carlsbad, California
| | | |
Collapse
|
23
|
Chawla S, Rahar B, Saxena S. S1P prophylaxis mitigates acute hypobaric hypoxia-induced molecular, biochemical, and metabolic disturbances: A preclinical report. IUBMB Life 2016; 68:365-75. [PMID: 26959531 DOI: 10.1002/iub.1489] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 02/13/2016] [Indexed: 12/17/2022]
Abstract
Sphingosine-1-phosphate (S1P) is emerging to have hypoxic preconditioning potential in various preclinical studies. The study aims to evaluate the preclinical preconditioning efficacy of exogenously administered S1P against acute hypobaric hypoxia (HH)-induced pathological disturbances. Male Sprague Dawley rats (200 ± 20 g) were preconditioned with 1, 10, and 100 μg/kg body weight (b.w.) S1P (i.v.) for three consecutive days. On the third day, S1P preconditioned animals, along with hypoxia control animals, were exposed to HH equivalent to 7,620 m (280 mm Hg) for 6 h. Postexposure status of cardiac energy production, circulatory vasoactive mediators, pulmonary and cerebral oxidative damage, and inflammation were assessed. HH exposure led to cardiac energy deficit indicated by low ATP levels and pronounced AMPK activation levels, raised circulatory levels of brain natriuretic peptide and endothelin-1 with respect to total nitrate (NOx), redox imbalance, inflammation, and alterations in NOx levels in the pulmonary and cerebral tissues. These pathological precursors have been routinely reported to be coincident with high-altitude diseases. Preconditioning with S1P, especially 1 µg/kg b.w. dose, was seen to reverse the manifestation of these pathological disturbances. The protective efficacy could be attributed, at least in part, to enhanced activity of cardioprotective protein kinase C and activation of small GTPase Rac1, which led to further induction of hypoxia-adaptive molecular mediators: hypoxia-inducible factor (HIF)-1α and Hsp70. This is a first such report, to the best of our knowledge, elucidating the mechanism of exogenous S1P-mediated HIF-1α/Hsp70 induction. Conclusively, systemic preconditioning with 1 μg/kg b.w. S1P in rats protects against acute HH-induced pathological disturbances. © 2016 IUBMB Life 68(5):365-375, 2016.
Collapse
Affiliation(s)
- Sonam Chawla
- Experimental Biology Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Timarpur, New Delhi, India
| | - Babita Rahar
- Experimental Biology Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Timarpur, New Delhi, India
| | - Shweta Saxena
- Experimental Biology Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Timarpur, New Delhi, India
| |
Collapse
|
24
|
Qi T, Xu F, Yan X, Li S, Li H. Sulforaphane exerts anti-inflammatory effects against lipopolysaccharide-induced acute lung injury in mice through the Nrf2/ARE pathway. Int J Mol Med 2015; 37:182-8. [PMID: 26531002 DOI: 10.3892/ijmm.2015.2396] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 10/16/2015] [Indexed: 11/06/2022] Open
Abstract
Sulforaphane (1-isothiocyanate-4-methyl sulfonyl butane) is a plant extract (obtained from cruciferous vegetables, such as broccoli and cabbage) and is known to exert anticancer, antioxidant and anti-inflammatory effects. It stimulates the generation of human or animal cells, which is beneficial to the body. The aim of the current study was to determine whether sulforaphane protects against lipopolysaccharide (LPS)‑induced acute lung injury (ALI) through its anti-inflammatory effects, and to investigate the signaling pathways involved. For this purpose, male BALB/c mice were treated with sulforaphane (50 mg/kg) and 3 days later, ALI was induced by the administration of LPS (5 mg/kg) and we thus established the model of ALI. Our results revealed that sulforaphane significantly decreased lactate dehydrogenase (LDH) activity (as shown by LDH assay), the wet-to-dry ratio of the lungs and the serum levels of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) (measured by ELISA), as well as nuclear factor-κB protein expression in mice with LPS-induced ALI. Moreover, treatment with sulforaphane significantly inhibited prostaglandin E2 (PGE2) production, and cyclooxygenase-2 (COX-2), matrix metalloproteinase-9 (MMP-9) protein expression (as shown by western blot analysis), as well as inducible nitric oxide synthase (iNOS) activity in mice with LPS-induced ALI. Lastly, we noted that pre-treatment with sulforaphane activated the nuclear factor-E2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway in the mice with LPS-induced ALI. These findings demonstrate that sulforaphane exerts protective effects against LPS-induced ALI through the Nrf2/ARE pathway. Thus, sulforaphane may be a potential a candidate for use in the treatment of ALI.
Collapse
Affiliation(s)
- Tianjie Qi
- Department of Respiratory Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Fei Xu
- Department of Cardiology, Jin Zhou People's Hospital of Hebei Province, Hebei 052260, P.R. China
| | - Xixin Yan
- Department of Respiratory Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Shuai Li
- Department of Respiratory Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Haitao Li
- Department of Respiratory Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
25
|
Zhu HT, Bian C, Yuan JC, Liao XJ, Liu W, Zhu G, Feng H, Lin JK. Hyperbaric oxygen therapy ameliorates acute brain injury after porcine intracerebral hemorrhage at high altitude. Crit Care 2015; 19:255. [PMID: 26073666 PMCID: PMC4522125 DOI: 10.1186/s13054-015-0976-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 06/04/2015] [Indexed: 11/10/2022] Open
Abstract
Introduction Intracerebral hemorrhage (ICH) at high altitude is not well understood to date. This study investigates the effects of high altitude on ICH, and examines the acute neuroprotection of hyperbaric oxygen (HBO) therapy against high-altitude ICH. Methods Minipigs were placed in a hypobaric chamber for 72 h before the operation. ICH was induced by an infusion of autologous arterial blood (3 ml) into the right basal ganglia. Animals in the high-altitude ICH group received HBO therapy (2.5 ATA for 60 min) 30 min after ICH. Blood gas, blood glucose and brain tissue oxygen partial pressure (PbtO2) were monitored continuously for animals from all groups, as were microdialysis products including glucose, lactate, pyruvate and glutamate in perihematomal tissue from 3 to 12 h post-ICH. Results High-altitude ICH animals showed significantly lower PbtO2, higher lactate/pyruvate ratio (LPR) and glutamate levels than low-altitude ICH animals. More severe neurological deficits, brain edema and neuronal damage were also observed in high-altitude ICH. After HBO therapy, PbtO2 was significantly increased and LPR and glutamate levels were significantly decreased. Brain edema, neurological deficits and neuronal damage were also ameliorated. Conclusions The data suggested a more serious disturbance of tissue oxygenation and cerebral metabolism in the acute stage after ICH at high altitude. Early HBO treatment reduced acute brain injury, perhaps through a mechanism involving the amelioration of the derangement of cerebral oxygenation and metabolism following high-altitude ICH.
Collapse
Affiliation(s)
- Hai-tao Zhu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 30 Gaotanyan Street, Chongqing, 400038, China.
| | - Chen Bian
- Department of Military Psychology, College of Psychology, Third Military Medical University, 30 Gaotanyan Street, Chongqing, 400038, China.
| | - Ji-chao Yuan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 30 Gaotanyan Street, Chongqing, 400038, China.
| | - Xiao-jun Liao
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 30 Gaotanyan Street, Chongqing, 400038, China.
| | - Wei Liu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 30 Gaotanyan Street, Chongqing, 400038, China.
| | - Gang Zhu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 30 Gaotanyan Street, Chongqing, 400038, China.
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 30 Gaotanyan Street, Chongqing, 400038, China.
| | - Jiang-kai Lin
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 30 Gaotanyan Street, Chongqing, 400038, China.
| |
Collapse
|
26
|
High-altitude pulmonary edema can be prevented by heat shock protein 70-mediated hyperbaric oxygen preconditioning. J Trauma Acute Care Surg 2015; 77:585-91. [PMID: 25250598 DOI: 10.1097/ta.0000000000000408] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The primary goal of this study was to test whether high-altitude exposure (HAE of 9.7% O2 at 0.47 absolute atmosphere [ATA] for 3 days) was capable of increasing lung edema, neutrophil, and hemorrhage scores as well as decreasing lung levels of both aquaporin 1 (AQP1) and AQP5 proteins and messenger RNA (mRNA) expression in rats, with a secondary goal to test whether a preinduction of heat shock protein 70 (HSP70) by hyperbaric oxygen preconditioning (HBO2P of 100% O2 at 2.0 ATA for 1 hour per day for 5 consecutive days) attenuated the HAE-induced increased lung injury scores and decreased lung AQP1 and AQP5 protein and mRNA expressions. METHODS Rats were assigned to (1) non-HBO2P (21% O2 at 1.0 ATA) + non-HAE (21% O2 at 1.0 ATA) group; (2) non-HBO2P + HAE group; (3) HBO2P + HAE group; and HBO2P + HSP70 antibodies (Ab) + HAE group. For the HSP70 Ab group, a neutralizing HSP70 Ab was injected intravenously at 24 hours before HAE. All the physiologic and biochemical parameters were obtained at the end of HAE or the equivalent period of non-HAE. The cardiovascular and blood gas parameters were monitored for all experiments. Bronchoalveolar lavage (BAL) was performed to determine proinflammatory cytokines (interleukin 6, interleukin 1β, and tumor necrosis factor α). Parts of the lung were excised for myeloperoxidase activity measurement, whereas the rest was collected for lung damage score assessments. AQP1 and AQP5 protein and mRAN expressions were also determined in the lung tissues. RESULTS In the non-HBO2P + HAE group, the animals displayed higher values of lung myeloperoxidase activity, BAL proinflammatory cytokines, lung water weight, and acute lung injury scores compared with those of the non-HBO2P + non-HAE controls. In contrast, the non-HBO2P + HAE group rats had lower values of lung AQP1 and AQP5 protein and mRNA expressions, mean arterial pressure, heart rate, SO2, Paco2, HCO3, and pH compared with those of non-HBO2P + non-HAE group rats. The increased acute lung edema, neutrophil, and hemorrhage scores; increased BAL levels of proinflammatory cytokines; decreased lung AQP1 and AQP5 protein and mRNA expressions; and hypotension, bradycardia, hypoxia, and acidosis caused by HAE were all significantly attenuated by HBO2P. CONCLUSION Our data indicate that HBO2P may attenuate high-altitude acute lung injury by a preinduction of lung HSP70 in rats.
Collapse
|
27
|
Chitra L, Boopathy R. Altered mitochondrial biogenesis and its fusion gene expression is involved in the high-altitude adaptation of rat lung. Respir Physiol Neurobiol 2013; 192:74-84. [PMID: 24361501 DOI: 10.1016/j.resp.2013.12.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 12/09/2013] [Accepted: 12/10/2013] [Indexed: 11/25/2022]
Abstract
Intermittent hypobaric hypoxia-induced preconditioning (IHH-PC) of rat favored the adaption of lungs to severe HH conditions, possibly through stabilization of mitochondrial function. This is based on the data generated on regulatory coordination of nuclear DNA-encoded mitochondrial biogenesis; dynamics, and mitochondrial DNA (mtDNA)-encoded oxidative phosphorylation (mtOXPHOS) genes expression. At 16th day after start of IHH-PC (equivalent to 5000m, 6h/d, 2w of treatment), rats were exposed to severe HH stimulation at 9142m for 6h. The IHH-PC significantly counteracted the HH-induced effect of increased lung: water content; tissue damage; and oxidant injury. Further, IHH-PC significantly increased the mitochondrial number, mtDNA content and mtOXPHOS complex activity in the lung tissues. This observation is due to an increased expression of genes involved in mitochondrial biogenesis (PGC-1α, ERRα, NRF1, NRF2 and TFAM), fusion (Mfn1 and Mfn2) and mtOXPHOS. Thus, the regulatory pathway formed by PGC-1α/ERRα/Mfn2 axes is required for the mitochondrial adaptation provoked by IHH-PC regimen to counteract subsequent HH stress.
Collapse
Affiliation(s)
- Loganathan Chitra
- Molecular Biology and Biotechnology Division, DRDO - BU Center for Life Sciences, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| | - Rathanam Boopathy
- Department of Biotechnology, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India.
| |
Collapse
|
28
|
Jain K, Suryakumar G, Prasad R, Ganju L. Upregulation of cytoprotective defense mechanisms and hypoxia-responsive proteins imparts tolerance to acute hypobaric hypoxia. High Alt Med Biol 2013; 14:65-77. [PMID: 23537263 DOI: 10.1089/ham.2012.1064] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Exposure to high altitude is a well-known environmental stress with physiological and metabolic consequences, with the major stressor being hypobaric hypoxia. The disruption in cellular homeostasis elicits several acute and chronic adaptations designed to diminish the stress imposed by the hypoxic insult. Highly conserved cellular machinery protects the myocardium from damage under reduced oxygen tension. In the present study, adult Sprague-Dawley rats were exposed to an altitude of 9754 m in a decompression chamber and screened on the basis of the time taken for onset of gasping. The animals were grouped as susceptible (<10 min), normal (10-25 min), and tolerant (>25 min). Histologically, susceptible animals showed increased myocardial inflammation and infiltration and greater CK-MB activity. These animals showed a three-fold increase in reactive oxygen species levels and subsequent oxidative damage to proteins and lipids as compared to control unexposed group. In tolerant animals, the damage was minimal. The resistance to damage in these animals was possibly due to enhanced myocardial antioxidant enzymes, catalase and superoxide dismutase. A significantly higher expression of HIF-1α and its responsive genes, including EPO, HO-1, and GLUT1, was seen in tolerant animals, although VEGF expression was enhanced in the susceptible group. Cytoprotective chaperones, HSP70 and HSP90, were elevated in the tolerant animals. The differential expression of these hypoxia-responsive molecules may thus act as potential biochemical markers for screening and identifying individuals susceptible to environmental stress.
Collapse
Affiliation(s)
- Kanika Jain
- Cellular Biochemistry Division, Defence Institute of Physiology and Allied Sciences, Timarpur, Delhi, India
| | | | | | | |
Collapse
|
29
|
Chitra L, Boopathy R. Adaptability to hypobaric hypoxia is facilitated through mitochondrial bioenergetics: an in vivo study. Br J Pharmacol 2013; 169:1035-47. [PMID: 23517027 PMCID: PMC3696327 DOI: 10.1111/bph.12179] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 02/22/2013] [Accepted: 03/06/2013] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND AND PURPOSE High-altitude pulmonary oedema (HAPE) experienced under high-altitude conditions is attributed to mitochondrial redox distress. Hence, hypobaric hypoxia (HH)-induced alteration in expression of mitochondrial biogenesis and dynamics genes was determined in rat lung. Further, such alteration was correlated with expression of mitochondrial DNA (mtDNA)-encoded oxidative phosphorylation (mtOXPHOS) genes. The prophylactic effect of dexamethasone (DEX) in counteracting the HH-induced mitochondrial distress was used as control to understand adaptation to high-altitude exposure. EXPERIMENTAL APPROACH Rats pretreated with DEX were exposed to normobaric normoxia (NN) or HH. HH-induced injury was assessed as an increase in lung water content, tissue damage and oxidant generation. Mitochondrial number, mtDNA content and mtOXPHOS activities were measured to determine mitochondrial function. The expression of mitochondrial biogenesis, dynamics and mtOXPHOS genes was studied. KEY RESULTS HH-induced lung injury was associated with decreased mitochondrial number, mtDNA content and mtOXPHOS activities. HH exposure decreased the nuclear gene oestrogen-related receptor-α (ERRα), which interacts with PPAR-γ coactivator-1α (PGC-1α) in controlling mitochondrial metabolism. Consequently, mtOXPHOS transcripts are repressed under HH. Further, HH modulated mitochondrial dynamics by decreasing mitofusin 2 (Mfn2) and augmenting fission 1 (Fis1) and dynamin-related protein 1 (Drp1) expression. Nevertheless, DEX treatment under NN (i.e. adaptation to HH) did not affect mitochondrial biogenesis and dynamics, but increased mtOXPHOS transcripts. Further, mtOXPHOS activities increased together with reduced oxidant generation. Also, DEX pretreatment normalized ERRα along with mitochondrial dynamics genes and increased mtOXPHOS transcripts to elicit the mitochondrial function under HH. CONCLUSIONS AND IMPLICATIONS HH stress (HAPE)-mediated mitochondrial dysfunction is due to repressed ERRα and mtOXPHOS transcripts. Thus, ERRα-mediated protection of mitochondrial bioenergetics might be the likely candidate required for lung adaptation to HH.
Collapse
Affiliation(s)
- Loganathan Chitra
- Molecular Biology and Biotechnology Division, DRDO – BU Center for Life Sciences, Bharathiar UniversityCoimbatore, India
| | | |
Collapse
|
30
|
Attenuating heatstroke-induced acute lung inflammation, edema, and injury in rats by exercise preconditioning. J Trauma Acute Care Surg 2013; 74:1052-9. [PMID: 23511144 DOI: 10.1097/ta.0b013e31827a3618] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND This study aimed to ascertain whether heat-induced acute lung edema, inflammation, and ischemic damage can be affected by heat shock protein 70 (HSP-70)-mediated exercise preconditioning (EP) in rats. METHODS Wistar rats were assigned to one of the following four groups: the non-EP + nonheated group, the non-EP + heated group, the EP + heated group, and the EP + HSP-70 antibodies + heated group. EP groups of animals were subjected to a protocol of running on a treadmill for 30 minutes at 20 m/min, 30 minutes at 30 m/min, and 60 minutes at 30 m/min after 1, 2, and 3 weeks of training, respectively. Heated group of animals, under general anesthesia, were put in a folded heating pad of 43°C for 68 minutes. Then, the heated animals were allowed to recover at room temperature. HSP-70 antibodies were injected intravenously 24 hours before heat exposure. RESULTS As compared with the non heated + non-EP rats, the heated + non-EP rats had significantly higher scores of alveolar edema, neutrophil infiltration, and hemorrhage, acute pleurisy, and increased bronchoalveolar fluid levels of proinflammatory cytokines and ischemic and oxidative damage markers. EP, in addition to inducing overexpression of HSP-70 in lung tissues, significantly attenuated heat-induced acute pulmonary edema, inflammation, and ischemic and oxidative damage in the lungs. HSP-70 antibodies, in addition to reducing HSP-70 expression in the lungs, significantly attenuated the beneficial effects of EP in reducing acute lung inflammation and injury. CONCLUSION EP may attenuate the occurrence of pulmonary edema, inflammation, as well as ischemic and oxidative damage caused by heatstroke by up-regulating HSP-70 in the lungs.
Collapse
|
31
|
Wang CT, Lin HJ, Cheng BC, Lin MT, Chang CP. Attenuating systemic inflammatory markers in simulated high-altitude exposure by heat shock protein 70-mediated hypobaric hypoxia preconditioning in rats. J Formos Med Assoc 2013; 114:328-38. [PMID: 25839766 DOI: 10.1016/j.jfma.2012.11.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Revised: 10/22/2012] [Accepted: 11/23/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND/PURPOSE The primary goal of this study was to test whether high-altitude exposure (HAE: 0.9% O(2) at 0.47 ATA for 24 hours) was capable of increasing the systemic inflammatory markers as well as the toxic organ injury indicators in rats, with a secondary goal to test whether preinduction of heat shock protein (HSP) 70 by hypobaric hypoxia preconditioning (HHP: 18.3% O(2) at 0.66 ATA for 5 h/day on 5 days consecutively for 2 weeks) attenuated the proposed increased serum levels of both the systemic inflammatory markers and the toxic organ injury indicators. METHODS Rats were assigned to: (1) non-HHP (21% O(2) at 1.0 ATA)+non-HAE (21% O(2) at 1.0 ATA) group; (2) non-HHP+HAE group; (3) HHP+non-HAE group; (4) HHP+HAE group; and (5) HHP+HSP70 antibodies (Ab)+HAE group. For the HSP70Ab group, a neutralizing HSP70Ab was injected intravenously at 24 hours prior to HAE. All the physiological and biochemical parameters were obtained at the end of HAE or the equivalent time period of non-HAE. Blood samples were obtained for determination of both the systemic inflammatory markers (e.g., serum tumor necrosis factor-α, interleukin-1β, E-selectin, intercellular adhesion molecule-1, and liver myeloperoxidase activity) and the toxic organ injury indicators (e.g., nitric oxide metabolites, 2,3-dihydroxybenzoic acid, and lactate dehydrogenase). RESULTS HHP, in addition to inducing overexpression of tissue HSP70, significantly attenuated the HAE-induced hypotension, bradycardia, hypoxia, acidosis, and increased tissue levels of both the systemic inflammatory markers and the toxic organ injury indicators. The beneficial effects of HHP in inducing tissue overexpression of HSP70 as well as in preventing the HAE-induced increased levels of the systemic inflammatory markers and the toxic organ injury indicators could be significantly reduced by HSP70Ab preconditioning. CONCLUSION These results suggest that HHP may downgrade both the systemic inflammatory markers and the toxic organ injury indicators in HAE by upregulating tissue HSP70.
Collapse
Affiliation(s)
- Chia-Ti Wang
- Department of Emergency Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Hung-Jung Lin
- Department of Emergency Medicine, Chi Mei Medical Center, Tainan, Taiwan; Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Bor-Chih Cheng
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan; Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan
| | - Mao-Tsun Lin
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Ching-Ping Chang
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan.
| |
Collapse
|
32
|
Wang LC, Chang CP, Chio CC, Wu MH, Lee YS, Huang CY, Tsai KJ. Hypobaric hypoxia preconditioning attenuates experimental heatstroke syndromes via preinduction of heat shock protein 70. Am J Med Sci 2012; 344:383-90. [PMID: 22245947 DOI: 10.1097/maj.0b013e31824314fe] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Heatstroke has been defined as a form of hyperthermia associated with a systemic inflammatory response that leads to multiple organ dysfunction syndrome (MODS). It has also been documented that heat shock protein 70 (HSP70) preconditioning is able to induce thermotolerance. Here, the authors further investigated whether hypobaric hypoxia preconditioning (HHP) improved the MODS in heatstroke by up-regulation of HSP70. METHODS Anesthetized rats were randomly assigned to (a) non-HHP + nonheated group, (b) non-HHP + heated group, (c) HHP + heated group and (d) HHP + HSP70 antibodies (Abs) + heated groups. All heated groups were exposed to heat stress (43°C, 70 minutes) to induce heatstroke. For HHP, animals were exposed to 0.66 atmosphere absolute (18.3% O2) for 5 hours daily for consecutive 5 days per week for 2 weeks before the start of heat exposure. RESULTS HHP significantly (i) attenuated hypotension, (ii) reduced plasma index of the toxic oxidizing radicals and the organ injury indicator, (iii) attenuated plasma systemic inflammatory response molecules, (iv) reduced an index of infiltration of polymorphonuclear neutrophils in the lung like myeloper-oxidase activity, (v) promoted plasma levels of an anti-inflammatory cytokine, interleukin-10, (vi) promoted the survival time to fourfold compared with non-HHP group and (vii) promoted the overexpression of HSP70 in different organs (eg, the lung) during heatstroke. The beneficial effects of HHP could be significantly attenuated by HSP70 Ab preconditioning. CONCLUSION Our results show that HHP protects rats from heat-induced MODS via up-regulating HSP70. Thus, HHP could be a novel strategy for the prevention of heatstroke animals or patients before heat exposure.
Collapse
Affiliation(s)
- Liang-Chao Wang
- Institute of Clinical Medicine, Department of Surgery, National Cheng-Kung University Hospital, Tainan, Taiwan
| | | | | | | | | | | | | |
Collapse
|
33
|
Lung oxidative damage by hypoxia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2012; 2012:856918. [PMID: 22966417 PMCID: PMC3433143 DOI: 10.1155/2012/856918] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 07/11/2012] [Indexed: 12/12/2022]
Abstract
One of the most important functions of lungs is to maintain an adequate oxygenation in the organism. This organ can be affected by hypoxia facing both physiological and pathological situations. Exposure to this condition favors the increase of reactive oxygen species from mitochondria, as from NADPH oxidase, xanthine oxidase/reductase, and nitric oxide synthase enzymes, as well as establishing an inflammatory process. In lungs, hypoxia also modifies the levels of antioxidant substances causing pulmonary oxidative damage. Imbalance of redox state in lungs induced by hypoxia has been suggested as a participant in the changes observed in lung function in the hypoxic context, such as hypoxic vasoconstriction and pulmonary edema, in addition to vascular remodeling and chronic pulmonary hypertension. In this work, experimental evidence that shows the implied mechanisms in pulmonary redox state by hypoxia is reviewed. Herein, studies of cultures of different lung cells and complete isolated lung and tests conducted in vivo in the different forms of hypoxia, conducted in both animal models and humans, are described.
Collapse
|
34
|
MO LIQIU, YANG CHUNTAO, GU MOFA, ZHENG DONGDAN, LIN LIN, WANG XIUYU, LAN AIPING, HU FEN, FENG JIANQIANG. PI3K/Akt signaling pathway-induced heme oxygenase-1 upregulation mediates the adaptive cytoprotection of hydrogen peroxide preconditioning against oxidative injury in PC12 cells. Int J Mol Med 2012; 30:314-20. [DOI: 10.3892/ijmm.2012.1002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 04/02/2012] [Indexed: 11/06/2022] Open
|
35
|
Attenuating brain edema, hippocampal oxidative stress, and cognitive dysfunction in rats using hyperbaric oxygen preconditioning during simulated high-altitude exposure. J Trauma Acute Care Surg 2012; 72:1220-7. [DOI: 10.1097/ta.0b013e318246ee70] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
36
|
Sightings edited by John W. Severinghaus. High Alt Med Biol 2012. [DOI: 10.1089/ham.2012.1312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
37
|
Sightings edited by John W. Severinghaus. High Alt Med Biol 2011. [DOI: 10.1089/ham.2011.1243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|